501
|
Pacheco-García U, Serafín-López J. Indirect Dispersion of SARS-CoV-2 Live-Attenuated Vaccine and Its Contribution to Herd Immunity. Vaccines (Basel) 2023; 11:655. [PMID: 36992239 PMCID: PMC10055900 DOI: 10.3390/vaccines11030655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
It has been 34 months since the beginning of the SARS-CoV-2 coronavirus pandemic, which causes the COVID-19 disease. In several countries, immunization has reached a proportion near what is required to reach herd immunity. Nevertheless, infections and re-infections have been observed even in vaccinated persons. That is because protection conferred by vaccines is not entirely effective against new virus variants. It is unknown how often booster vaccines will be necessary to maintain a good level of protective immunity. Furthermore, many individuals refuse vaccination, and in developing countries, a large proportion of the population has not yet been vaccinated. Some live-attenuated vaccines against SARS-CoV-2 are being developed. Here, we analyze the indirect dispersion of a live-attenuated virus from vaccinated individuals to their contacts and the contribution that this phenomenon could have to reaching Herd Immunity.
Collapse
Affiliation(s)
- Ursino Pacheco-García
- Department of Cardio-Renal Pathophysiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico City 14080, Mexico
| | - Jeanet Serafín-López
- Department of Immunology, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico
| |
Collapse
|
502
|
McCollum C, Courtney CM, O’Connor NJ, Aunins TR, Jordan TX, Rogers KL, Brindley S, Brown JM, Nagpal P, Chatterjee A. Safety and Biodistribution of Nanoligomers Targeting the SARS-CoV-2 Genome for the Treatment of COVID-19. ACS Biomater Sci Eng 2023; 9:1656-1671. [PMID: 36853144 PMCID: PMC10000012 DOI: 10.1021/acsbiomaterials.2c00669] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023]
Abstract
As the world braces to enter its fourth year of the coronavirus disease 2019 (COVID-19) pandemic, the need for accessible and effective antiviral therapeutics continues to be felt globally. The recent surge of Omicron variant cases has demonstrated that vaccination and prevention alone cannot quell the spread of highly transmissible variants. A safe and nontoxic therapeutic with an adaptable design to respond to the emergence of new variants is critical for transitioning to the treatment of COVID-19 as an endemic disease. Here, we present a novel compound, called SBCoV202, that specifically and tightly binds the translation initiation site of RNA-dependent RNA polymerase within the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome, inhibiting viral replication. SBCoV202 is a Nanoligomer, a molecule that includes peptide nucleic acid sequences capable of binding viral RNA with single-base-pair specificity to accurately target the viral genome. The compound has been shown to be safe and nontoxic in mice, with favorable biodistribution, and has shown efficacy against SARS-CoV-2 in vitro. Safety and biodistribution were assessed using three separate administration methods, namely, intranasal, intravenous, and intraperitoneal. Safety studies showed the Nanoligomer caused no outward distress, immunogenicity, or organ tissue damage, measured through observation of behavior and body weight, serum levels of cytokines, and histopathology of fixed tissue, respectively. SBCoV202 was evenly biodistributed throughout the body, with most tissues measuring Nanoligomer concentrations well above the compound KD of 3.37 nM. In addition to favorable availability to organs such as the lungs, lymph nodes, liver, and spleen, the compound circulated through the blood and was rapidly cleared through the renal and urinary systems. The favorable biodistribution and lack of immunogenicity and toxicity set Nanoligomers apart from other antisense therapies, while the adaptability of the nucleic acid sequence of Nanoligomers provides a defense against future emergence of drug resistance, making these molecules an attractive potential treatment for COVID-19.
Collapse
Affiliation(s)
- Colleen
R. McCollum
- Department
of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - Colleen M. Courtney
- Department
of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80303, United States
- Sachi Bio, Colorado Technology Center, Louisville, Colorado 80027, United States
| | - Nolan J. O’Connor
- Department
of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - Thomas R. Aunins
- Department
of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - Tristan X. Jordan
- Department
of Microbiology, New York University Langone, New York, New York 10016, United States
| | - Keegan L. Rogers
- Department
of Pharmaceutical Sciences, University of
Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Stephen Brindley
- Department
of Pharmaceutical Sciences, University of
Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Jared M. Brown
- Department
of Pharmaceutical Sciences, University of
Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Prashant Nagpal
- Sachi Bio, Colorado Technology Center, Louisville, Colorado 80027, United States
- Antimicrobial
Regeneration Consortium Labs, Louisville, Colorado 80027, United States
| | - Anushree Chatterjee
- Department
of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80303, United States
- Sachi Bio, Colorado Technology Center, Louisville, Colorado 80027, United States
- Antimicrobial
Regeneration Consortium Labs, Louisville, Colorado 80027, United States
| |
Collapse
|
503
|
Benkeser D, Fong Y, Janes HE, Kelly EJ, Hirsch I, Sproule S, Stanley AM, Maaske J, Villafana T, Houchens CR, Martins K, Jayashankar L, Castellino F, Ayala V, Petropoulos CJ, Leith A, Haugaard D, Webb B, Lu Y, Yu C, Borate B, van der Laan LWP, Hejazi NS, Carpp LN, Randhawa AK, Andrasik MP, Kublin JG, Isaacs MB, Makhene M, Tong T, Robb ML, Corey L, Neuzil KM, Follmann D, Hoffman C, Falsey AR, Sobieszczyk M, Koup RA, Donis RO, Gilbert PB. Immune correlates analysis of a phase 3 trial of the AZD1222 (ChAdOx1 nCoV-19) vaccine. NPJ Vaccines 2023; 8:36. [PMID: 36899062 PMCID: PMC10005913 DOI: 10.1038/s41541-023-00630-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
In the phase 3 trial of the AZD1222 (ChAdOx1 nCoV-19) vaccine conducted in the U.S., Chile, and Peru, anti-spike binding IgG concentration (spike IgG) and pseudovirus 50% neutralizing antibody titer (nAb ID50) measured four weeks after two doses were assessed as correlates of risk and protection against PCR-confirmed symptomatic SARS-CoV-2 infection (COVID-19). These analyses of SARS-CoV-2 negative participants were based on case-cohort sampling of vaccine recipients (33 COVID-19 cases by 4 months post dose two, 463 non-cases). The adjusted hazard ratio of COVID-19 was 0.32 (95% CI: 0.14, 0.76) per 10-fold increase in spike IgG concentration and 0.28 (0.10, 0.77) per 10-fold increase in nAb ID50 titer. At nAb ID50 below the limit of detection (< 2.612 IU50/ml), 10, 100, and 270 IU50/ml, vaccine efficacy was -5.8% (-651%, 75.6%), 64.9% (56.4%, 86.9%), 90.0% (55.8%, 97.6%) and 94.2% (69.4%, 99.1%). These findings provide further evidence towards defining an immune marker correlate of protection to help guide regulatory/approval decisions for COVID-19 vaccines.
Collapse
Affiliation(s)
- David Benkeser
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Holly E Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Elizabeth J Kelly
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ian Hirsch
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Stephanie Sproule
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ann Marie Stanley
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Jill Maaske
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Tonya Villafana
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Christopher R Houchens
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - Karen Martins
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - Lakshmi Jayashankar
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - Flora Castellino
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - Victor Ayala
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | | | | | | | | | - Yiwen Lu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Chenchen Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Bhavesh Borate
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lars W P van der Laan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Nima S Hejazi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - April K Randhawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michele P Andrasik
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Mamodikoe Makhene
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tina Tong
- Vaccine Translational Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Merlin L Robb
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Lawrence Corey
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Corey Hoffman
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - Ann R Falsey
- Division of Infectious Diseases, Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Magdalena Sobieszczyk
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center and New York-Presbyterian Hospital, New York, NY, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ruben O Donis
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, Department of Health and Human Services, Washington, DC, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
504
|
Wankhede D, Grover S, Hofman P. Determinants of humoral immune response to SARS-CoV-2 vaccines in solid cancer patients: A systematic review and meta-analysis. Vaccine 2023; 41:1791-1798. [PMID: 36792435 PMCID: PMC9922575 DOI: 10.1016/j.vaccine.2023.01.072] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/15/2023]
Abstract
IMPORTANCE Solid cancer patients following SARS-CoV-2 vaccination are likely to have a lower seroconversion rate than healthy adults. Seroconversion between those with and without cancer is likely to vary moderately or to be restricted to specific subgroups. Therefore, we sought to conduct a systematic review and meta-analysis to identify risk factors for diminished humoral immune responses in solid cancer patients. METHODS MEDLINE, Embase, Web of Science, Cochrane Library, and ClinicalTrials.gov were used to search literature through May 1, 2022. Prospective or retrospective studies comparing responders with non-responders against SARS-CoV-2 spike (S) protein receptor-binding domain (RBD) following COVID-19 vaccination were included. Pooled Odds Ratios (pORs) with 95% CIs for binary variables and differences in means (with SDs) for continuous variables were calculated to determine the pooled effect estimates of risk factors for poor antibody response. RESULTS Fifteen studies enrolling 3593 patients were included in the analysis. Seroconversion was seen in 84% of the pooled study population. Male gender, age >65 years, and recent chemotherapy were all factors in a poor immune response. Patients under follow-up, those who received immunotherapy or targeted therapy, were more likely to be seropositive. Cancer subtypes, vaccine types, and timing of antibody testing from the 2nd dose of vaccine did not correlate with seroconversion. CONCLUSION Cytotoxic therapy for solid cancer may portend poor immune response following 2 doses of COVID-19 vaccines suggesting a need for booster doses in these patients. Immunotherapy and targeted therapy are likely to be associated with seropositive status, and thus can be considered as an alternative to cytotoxic agents in cases where both therapies are equally efficacious.
Collapse
Affiliation(s)
- Durgesh Wankhede
- Department of Surgical Oncology, All India Institute of Medical Sciences, New Delhi, India.
| | - Sandeep Grover
- Center for Human Genetics, Universitatsklinikum Giessen und Marburg - Standort Marburg, 35055 Marburg, Germany
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Côte d’Azur, 30 avenue de la voie romaine, 06002 Nice, France,Institute for Research on Cancer and Ageing, Nice (IRCAN), INSERM U1081 and UMR CNRS 7284, Team 4, Nice, France,Hospital-Integrated Biobank BB-0033-00025, Pasteur Hospital, Nice, France,University Hospital Federation OncoAge, CHU de Nice, University Côte d’Azur, Nice, France
| |
Collapse
|
505
|
de Sousa Neto AR, de Carvalho ARB, Ferreira da Silva MD, Rêgo Neta MM, Sena IVDO, Almeida RN, Filha FSSC, Lima e Silva LL, da Costa GR, Lira IMDS, Portela DMMC, Oliveira e Silva AT, Rabêlo CBDM, Valle ARMDC, Moura MEB, de Freitas DRJ. Bibliometric Analysis of Global Scientific Production on COVID-19 and Vaccines. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4796. [PMID: 36981704 PMCID: PMC10049169 DOI: 10.3390/ijerph20064796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 06/18/2023]
Abstract
This bibliometric analysis aims to analyze the global scientific production of COVID-19 and vaccines. First, a search for scientific articles was performed using the advanced query in the Web of Science™ database, more precisely in its core collection, on 18 February 2023. Data from 7754 articles were analyzed using the Bibliometrix R package and the Biblioshiny application. The evaluated articles were published mainly in 2022 (60%). The scientific journals that published the most about COVID-19 and vaccines were "Vaccines", "Vaccine" and "Human Vaccines & Immunotherapeutics". The University of Oxford was the most productive institution, with the authors of the articles mainly originating from the United States, China and the United Kingdom. The United States, despite having carried out the most significant number of collaborations, published mainly with local researchers. The 15 most cited articles and the KeyWords Plus™ evidenced the focus of the published articles on the safety and efficacy of vaccines against COVID-19, as well as on the evaluation of vaccine acceptance, more specifically on vaccine hesitancy. Research funding came primarily from US government agencies.
Collapse
|
506
|
Crombé A, Bensid L, Seux M, Fadli D, Arnaud F, Benhamed A, Banaste N, Gorincour G. Impact of Vaccination and the Omicron Variant on COVID-19-related Chest CT Findings: A Multicenter Study. Radiology 2023; 307:e222730. [PMID: 36880948 PMCID: PMC10031570 DOI: 10.1148/radiol.222730] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) omicron variant has a higher infection rate than previous variants but results in less severe disease. However, the impacts of omicron and vaccination on chest CT findings are difficult to evaluate. Purpose To investigate the impact of vaccination status and predominant variant on chest CT findings, diagnostic and severity scores in multicenter sample of consecutive patients referred to emergency departments for proven COVID-19. Materials and Methods This retrospective, multicenter study included adults referred to 93 emergency departments with SARS-CoV-2 infection according to RT-PCR and known vaccination status between July 2021 and March 2022. Clinical data and structured chest CT reports including semiquantitative diagnostic and severity scores following the French Society of Radiology-Thoracic Imaging Society guidelines were extracted from a teleradiology database. Observations were divided into 'delta-predominant', 'transition', and 'omicron-predominant' periods. Associations between scores and variant and vaccination status were investigated with Chi-square tests and ordinal regressions. Multivariable analyses evaluated the influence of omicron variant and vaccination status on the diagnostic and severity scores. Results Overall, 3876 patients were included (median age: 68 years [Q1-Q3: 54-80], 1695 females). Diagnostic and severity scores were associated with the predominant variant (delta- versus omicron-predominant, Chi-square=112.4 and 33.7, both P<.001) and vaccination (Chi-square=243.6 and 210, both P<.001) and their interaction (Chi-square=4.3, P=.04 and Chi-square=28.7, P<.001, respectively). In multivariable analyses, omicron variant was associated with lower odds of typical CT findings than delta variant (OR=0.46, P<.001). Two and three vaccine doses were associated with lower odds of demonstrating typical CT findings (OR=0.32 and OR=0.20, both P<.001), and of having high severity score (OR=0.47 and OR=0.33, both P<.001), compared with unvaccinated patients. Conclusion Both the omicron variant and vaccination were associated with less typical chest CT manifestations for COVID-19 and lesser extent of disease. See also the editorial by Yoon and Goo in this issue.
Collapse
Affiliation(s)
- Amandine Crombé
- IMADIS, 48 Rue Quivogne, Lyon, Bordeaux, Marseille, France
- Department of radiology, Pellegrin university hospital and Bordeaux
university, Bordeaux, France
| | - Lounès Bensid
- IMADIS, 48 Rue Quivogne, Lyon, Bordeaux, Marseille, France
| | - Mylène Seux
- IMADIS, 48 Rue Quivogne, Lyon, Bordeaux, Marseille, France
| | - David Fadli
- IMADIS, 48 Rue Quivogne, Lyon, Bordeaux, Marseille, France
- Department of radiology, Pellegrin university hospital and Bordeaux
university, Bordeaux, France
| | - François Arnaud
- IMADIS, 48 Rue Quivogne, Lyon, Bordeaux, Marseille, France
- Ramsay Générale de Santé, Hôpital
privé Clairval, Marseille, France
| | - Axel Benhamed
- Service SAMU-Urgences, Centre Hospitalier Universitaire
Édouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Nathan Banaste
- IMADIS, 48 Rue Quivogne, Lyon, Bordeaux, Marseille, France
- Ramsay Générale de Santé, Clinique Convert,
Bourg-en-Bresse
| | - Guillaume Gorincour
- IMADIS, 48 Rue Quivogne, Lyon, Bordeaux, Marseille, France
- ELSAN, Clinique Bouchard, Marseille, France
| |
Collapse
|
507
|
Zirkenbach VA, Ignatz RM, Öttl R, Cehreli Z, Stroikova V, Kaya M, Lehmann LH, Preusch MR, Frey N, Kaya Z. Effect of SARS-CoV-2 mRNA-Vaccine on the Induction of Myocarditis in Different Murine Animal Models. Int J Mol Sci 2023; 24:ijms24055011. [PMID: 36902442 PMCID: PMC10002951 DOI: 10.3390/ijms24055011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/02/2023] [Accepted: 03/04/2023] [Indexed: 03/08/2023] Open
Abstract
In the course of the SARS-CoV-2 pandemic, vaccination safety and risk factors of SARS-CoV-2 mRNA-vaccines were under consideration after case reports of vaccine-related side effects, such as myocarditis, which were mostly described in young men. However, there is almost no data on the risk and safety of vaccination, especially in patients who are already diagnosed with acute/chronic (autoimmune) myocarditis from other causes, such as viral infections, or as a side effect of medication and treatment. Thus, the risk and safety of these vaccines, in combination with other therapies that could induce myocarditis (e.g., immune checkpoint inhibitor (ICI) therapy), are still poorly assessable. Therefore, vaccine safety, with respect to worsening myocardial inflammation and myocardial function, was studied in an animal model of experimentally induced autoimmune myocarditis. Furthermore, it is known that ICI treatment (e.g., antibodies (abs) against PD-1, PD-L1, and CTLA-4, or a combination of those) plays an important role in the treatment of oncological patients. However, it is also known that treatment with ICIs can induce severe, life-threatening myocarditis in some patients. Genetically different A/J (most susceptible strain) and C57BL/6 (resistant strain) mice, with diverse susceptibilities for induction of experimental autoimmune myocarditis (EAM) at various age and gender, were vaccinated twice with SARS-CoV-2 mRNA-vaccine. In an additional A/J group, an autoimmune myocarditis was induced. In regard to ICIs, we tested the safety of SARS-CoV-2 vaccination in PD-1-/- mice alone, and in combination with CTLA-4 abs. Our results showed no adverse effects related to inflammation and heart function after mRNA-vaccination, independent of age, gender, and in different mouse strains susceptible for induction of experimental myocarditis. Moreover, there was no worsening effect on inflammation and cardiac function when EAM in susceptible mice was induced. However, in the experiments with vaccination and ICI treatment, we observed, in some mice, low elevation of cardiac troponins in sera, and low scores of myocardial inflammation. In sum, mRNA-vaccines are safe in a model of experimentally induced autoimmune myocarditis, but patients undergoing ICI therapy should be closely monitored when vaccinated.
Collapse
Affiliation(s)
| | - Rebecca M. Ignatz
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Renate Öttl
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Zeynep Cehreli
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Vera Stroikova
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Mansur Kaya
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Lorenz H. Lehmann
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
| | - Michael R. Preusch
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
| | - Ziya Kaya
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221-5639617
| |
Collapse
|
508
|
Stiepel RT, Duggan E, Batty CJ, Ainslie KM. Micro and nanotechnologies: The little formulations that could. Bioeng Transl Med 2023; 8:e10421. [PMID: 36925714 PMCID: PMC10013823 DOI: 10.1002/btm2.10421] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/22/2022] [Accepted: 09/18/2022] [Indexed: 11/05/2022] Open
Abstract
The first publication of micro- and nanotechnology in medicine was in 1798 with the use of the Cowpox virus by Edward Jenner as an attenuated vaccine against Smallpox. Since then, there has been an explosion of micro- and nanotechnologies for medical applications. The breadth of these micro- and nanotechnologies is discussed in this piece, presenting the date of their first report and their latest progression (e.g., clinical trials, FDA approval). This includes successes such as the recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines from Pfizer, Moderna, and Janssen (Johnson & Johnson) as well as the most popular nanoparticle therapy, liposomal Doxil. However, the enormity of the success of these platforms has not been without challenges. For example, we discuss why the production of Doxil was halted for several years, and the bankruptcy of BIND therapeutics, which relied on a nanoparticle drug carrier. Overall, the field of micro- and nanotechnology has advanced beyond these challenges and continues advancing new and novel platforms that have transformed therapies, vaccines, and imaging. In this review, a wide range of biomedical micro- and nanotechnology is discussed to serve as a primer to the field and provide an accessible summary of clinically relevant micro- and nanotechnology platforms.
Collapse
Affiliation(s)
- Rebeca T. Stiepel
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Eliza Duggan
- North Carolina School of Science and MathematicsDurhamNorth CarolinaUSA
| | - Cole J. Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North CarolinaChapel HillNorth CarolinaUSA
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill and North Carolina State UniversityChapel HillNorth CarolinaUSA
- Department of Microbiology and Immunology, UNC School of MedicineUniversity of North CarolinaChapel HillNorth CarolinaUSA
| |
Collapse
|
509
|
Giesen N, Busch E, Schalk E, Beutel G, Rüthrich MM, Hentrich M, Hertenstein B, Hirsch HH, Karthaus M, Khodamoradi Y, Koehler P, Krüger W, Koldehoff M, Krause R, Mellinghoff SC, Penack O, Sandherr M, Seggewiss-Bernhardt R, Spiekermann K, Sprute R, Stemler J, Weissinger F, Wörmann B, Wolf HH, Cornely OA, Rieger CT, von Lilienfeld-Toal M. AGIHO guideline on evidence-based management of COVID-19 in cancer patients: 2022 update on vaccination, pharmacological prophylaxis and therapy in light of the omicron variants. Eur J Cancer 2023; 181:102-118. [PMID: 36652889 PMCID: PMC9737523 DOI: 10.1016/j.ejca.2022.11.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022]
Abstract
The novel coronavirus SARS-CoV-2 and the associated infectious disease COVID-19 pose a significant challenge to healthcare systems worldwide. Patients with cancer have been identified as a high-risk population for severe infections, rendering prophylaxis and treatment strategies for these patients particularly important. Rapidly evolving clinical research, resulting in the recent advent of various vaccines and therapeutic agents against COVID-19, offers new options to improve care and protection of cancer patients. However, ongoing epidemiological changes and rise of new virus variants require repeated revisions and adaptations of prophylaxis and treatment strategies to meet these new challenges. Therefore, this guideline provides an update on evidence-based recommendations with regard to vaccination, pharmacological prophylaxis and treatment of COVID-19 in cancer patients in light of the currently dominant omicron variants. It was developed by an expert panel of the Infectious Diseases Working Party (AGIHO) of the German Society for Hematology and Medical Oncology (DGHO) based on a critical review of the most recent available data.
Collapse
Affiliation(s)
- Nicola Giesen
- Department of Hematology, Oncology and Palliative Care, Robert Bosch Hospital, Stuttgart, Germany.
| | - Elena Busch
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Enrico Schalk
- Department of Hematology and Oncology, Medical Centre, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Gernot Beutel
- Department for Haematology, Haemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany; Working Party Intensive Care in Haematologic and Oncologic Patients (iCHOP) of the German Society of Haematology and Medical Oncology (DGHO), Germany
| | - Maria M Rüthrich
- Department of Interdisciplinary Intensive Care Medicine, Vivantes Humboldt-Klinikum, Berlin, Germany
| | - Marcus Hentrich
- Department of Hematology and Oncology, Red Cross Hospital Munich, Munich, Germany
| | | | - Hans H Hirsch
- Transplantation & Clinical Virology, Department Biomedicine (Haus Petersplatz), University of Basel, Basel, Switzerland; Clinical Virology, Laboratory Medicine, University Hospital Basel, Basel, Switzerland; Infectious Diseases & Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - Meinolf Karthaus
- Department of Hematology, Oncology and Palliative Care, Klinikum Neuperlach/Klinikum Harlaching, Munich, Germany
| | - Yascha Khodamoradi
- Department of Internal Medicine, Infectious Diseases, Goethe University Frankfurt, Frankfurt Am Main, Germany
| | - Philipp Koehler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany
| | - William Krüger
- Department of Hematology and Oncology, Stem Cell Transplantation, Palliative Care, University Hospital Greifswald, Greifswald, Germany
| | - Michael Koldehoff
- Department of Bone Marrow Transplantation, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany; Department of Hygiene and Environmental Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Robert Krause
- Division of Infectious Diseases, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Sibylle C Mellinghoff
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Olaf Penack
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hematology, Oncology and Tumorimmunology, Berlin, Germany
| | - Michael Sandherr
- MVZ Penzberg, Department of Hematology and Oncology, Weilheim, Germany
| | - Ruth Seggewiss-Bernhardt
- Medizinische Klinik V, Sozialstiftung Bamberg, Bamberg, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Karsten Spiekermann
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Rosanne Sprute
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Jannik Stemler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Florian Weissinger
- Department of Internal Medicine, Hematology, Oncology, Stem Cell Transplantation and Palliative Care, Evangelisches Klinikum Bethel, Bielefeld, Germany
| | - Bernhard Wörmann
- Division of Haematology, Oncology and Tumor Immunology, Department of Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hans-Heinrich Wolf
- Department of Hematology, Oncology and Hemostaseology, Südharzklinikum Nordhausen, Nordhausen, Germany
| | - Oliver A Cornely
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), Cologne, Germany
| | - Christina T Rieger
- Hemato-Oncology Germering & Interdisciplinary Tumorcenter, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Marie von Lilienfeld-Toal
- Department of Haematology and Medical Oncology, Clinic for Internal Medicine II, University Hospital Jena, Jena, Germany; Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| |
Collapse
|
510
|
Khiali S, Rezagholizadeh A, Behzad H, Bannazadeh Baghi H, Entezari-Maleki T. Current evidence of COVID-19 vaccination-related cardiovascular events. Postgrad Med 2023; 135:102-120. [PMID: 36567602 DOI: 10.1080/00325481.2022.2161249] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Currently, the world is recovering from the shock of the coronavirus disease 2019 (COVID-19) pandemic; however, this situation is still fragile. Health authorities recommend administering COVID-19 vaccines as the safest and most reliable tool for eliminating COVID-19. Subsequent to the extensive administration of the COVID-19 vaccines, a series of cardiovascular adverse effects have been reported. This comprehensive review aimed to provide an update on the etiology, pathophysiology, clinical features, and management of the cardiovascular adverse events associated with COVID-19 vaccines, including myocarditis, pericarditis, thrombosis with thrombocytopenia syndrome, myocardial infarction, cardiac arrhythmias, hypertension, and stress-induced cardiomyopathy. The benefits of COVID-19 vaccination far outweigh the reported adverse events. It would be clinically important to provide diagnostic scoring systems to differentiate COVID-19-related cardiovascular adverse events from other causes and develop therapeutic approaches for their management. Further evaluation of cardiovascular adverse events of the COVID-19 vaccines is crucial for implementing vaccination programs and developing safer and more reliable vaccines.
Collapse
Affiliation(s)
- Sajad Khiali
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afra Rezagholizadeh
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Behzad
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Taher Entezari-Maleki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
511
|
Kodadek LM, Moore MS, Miller SM, Schneider EB, Ahuja V, Maerz LL, Davis KA. Palliative Care in a Pandemic: A Multicenter Cohort of Critically Ill Patients with Coronavirus Disease 2019. Surg Infect (Larchmt) 2023; 24:190-198. [PMID: 36757283 PMCID: PMC10081705 DOI: 10.1089/sur.2022.377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
Background: Trends in mortality, palliative care, and end-of-life care among critically ill patients with coronavirus disease 2019 (COVID-19) remain underreported. We hypothesized that use of palliative care and end-of-life care would increase over time, because improved understanding of the disease course and prognosis would potentially lead to more frequent use of these services. Patients and Methods: Adult patients with severe acute respiratory syndrome coronavirus 2 infection (SARS-CoV-2) during pandemic wave one (W1: March 2020 to September 2020) or wave two (W2: October 2020 to June 2021) admitted to an intensive care unit (ICU) in one of six northeastern U.S. hospitals were identified and clinical characteristics obtained. Vaccination data were unavailable. Outcomes of interest included mortality, palliative care consultation, and any end-of-life care (including hospice and comfort care). Results: There were 1,904 critically ill patients with COVID-19: 817 (42.9%) in W1 and 1,087 (57.1%) in W2. Patients received mechanical ventilation more often during W1 than W2 (52.9% vs. 46.3%; p = 0.004), with no difference in ICU or hospital length of stay between waves. Mortality between W1 and W2 was similar (31.2% vs. 30.9%; p = 0.888). There was no difference in use of palliative care or any end-of-life care between waves. Patients who died during W2 versus W1 were more likely to have received both mechanical ventilation (77.1% vs. 67.1%; p = 0.007) and palliative care services (52.1% vs. 41.2%; p = 0.009). However, logistic regression adjusted for demographics, baseline comorbid disease, and clinical characteristics showed no difference in mortality (odds ratio [OR], 1.15; 95% confidence interval [CI], 0.89-1.48), palliative care (OR, 1.08; 95% CI, 0.84-1.40), or any end-of-life care (OR, 1.05; 95% CI, 0.82-1.34) in W2 versus W1. Conclusions: Mortality among critically ill patients with COVID-19 has remained constant across two pandemic waves with no change in use of palliative or end-of-life care.
Collapse
Affiliation(s)
- Lisa M. Kodadek
- Division of General Surgery, Trauma, and Surgical Critical Care, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Miranda S. Moore
- Center for Health Services and Outcomes Research, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Eric B. Schneider
- Center for Health Services and Outcomes Research, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Vanita Ahuja
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
- Connecticut Healthcare System Veterans Affairs, West Haven, Connecticut, USA
| | - Linda L. Maerz
- Division of General Surgery, Trauma, and Surgical Critical Care, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kimberly A. Davis
- Division of General Surgery, Trauma, and Surgical Critical Care, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
512
|
Response to the letter to the Editor regarding the article “Vaccine versus infection - COVID-19-related loss of training time in elite athletes”. J Sci Med Sport 2023; 26:181-182. [PMID: 37061291 PMCID: PMC9987613 DOI: 10.1016/j.jsams.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
|
513
|
Ge J, Digitale JC, Pletcher MJ, Lai JC. Breakthrough SARS-CoV-2 infection outcomes in vaccinated patients with chronic liver disease and cirrhosis: A National COVID Cohort Collaborative study. Hepatology 2023; 77:834-850. [PMID: 36799617 PMCID: PMC9538384 DOI: 10.1002/hep.32780] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND AIMS Outcomes of breakthrough SARS-CoV-2 infections have not been well characterized in non-veteran vaccinated patients with chronic liver diseases (CLD). We used the National COVID Cohort Collaborative (N3C) to describe these outcomes. APPROACH AND RESULTS We identified all CLD patients with or without cirrhosis who had SARS-CoV-2 testing in the N3C Data Enclave as of January 15, 2022. We used Poisson regression to estimate incidence rates of breakthrough infections and Cox survival analyses to associate vaccination status with all-cause mortality at 30 days among infected CLD patients. We isolated 278,457 total CLD patients: 43,079 (15%) vaccinated and 235,378 (85%) unvaccinated. Of 43,079 vaccinated patients, 32,838 (76%) were without cirrhosis and 10,441 (24%) with cirrhosis. Breakthrough infection incidences were 5.4 and 4.9 per 1000 person-months for fully vaccinated CLD patients without cirrhosis and with cirrhosis, respectively. Of the 68,048 unvaccinated and 10,441 vaccinated CLD patients with cirrhosis, 15% and 3.7%, respectively, developed SARS-CoV-2 infection. The 30-day outcome of mechanical ventilation or death after SARS-CoV-2 infection for unvaccinated and vaccinated CLD patients with cirrhosis were 15.2% and 7.7%, respectively. Compared to unvaccinated patients with cirrhosis, full vaccination was associated with a 0.34-times adjusted hazard of death at 30 days. CONCLUSIONS In this N3C study, breakthrough infection rates were similar among CLD patients with and without cirrhosis. Full vaccination was associated with a 66% reduction in risk of all-cause mortality for breakthrough infection among CLD patients with cirrhosis. These results provide an additional impetus for increasing vaccination uptake in CLD populations.
Collapse
Affiliation(s)
- Jin Ge
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California–San Francisco, San Francisco, California, USA
| | - Jean C. Digitale
- Department of Epidemiology and Biostatistics, University of California–San Francisco, San Francisco, California, USA
| | - Mark J. Pletcher
- Department of Epidemiology and Biostatistics, University of California–San Francisco, San Francisco, California, USA
- Division of General Internal Medicine, Department of Medicine, University of California–San Francisco, San Francisco, California, USA
| | - Jennifer C. Lai
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California–San Francisco, San Francisco, California, USA
| |
Collapse
|
514
|
Wu PC, Li CL, Chang YT, Chen CC, Wu CY, Ma SH. Management of Atopic Dermatitis During the COVID-19 Pandemic: Key Questions and Review of the Current Evidence. Dermatitis 2023; 34:77-84. [PMID: 36917535 DOI: 10.1089/derm.2022.29019.pwu] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Since the outbreak of COVID-19, management of atopic dermatitis (AD) has been widely discussed. Key issues include the risk of COVID-19 infection and related outcomes in AD patients, the efficacy and safety of COVID-19 vaccination in AD populations, and management of AD in the COVID-19 pandemic. Recent studies have shown that patients with AD have a slightly increased risk of COVID-19 infection but are not associated with a worse outcome than the non-AD population. COVID-19 vaccination is generally effective and safe in patients with AD. However, temporary discontinuation of certain systemic immunomodulatory agents after vaccination is suggested. During the pandemic, continuation of all immunomodulating agents is suggested, but these agents should be paused when patients with AD are infected with COVID-19 until recovery. Further studies are warranted to investigate the long-term interaction between AD and COVID-19 to aid clinical decisions during the pandemic.
Collapse
Affiliation(s)
- Po-Chien Wu
- From the *Department of Dermatology, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
- Research Center of Big Data and Meta-Analysis, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan
| | - Chia-Lun Li
- Department of Dermatology, Taipei Veterans General Hospital, Taipei City, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yun-Ting Chang
- Department of Dermatology, Taipei Veterans General Hospital, Taipei City, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dermatology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Chiang Chen
- Department of Dermatology, Taipei Veterans General Hospital, Taipei City, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dermatology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chen-Yi Wu
- Department of Dermatology, Taipei Veterans General Hospital, Taipei City, Taiwan
- Department of Dermatology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Public Health, Institute of Public Health, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Sheng-Hsiang Ma
- Department of Dermatology, Taipei Veterans General Hospital, Taipei City, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
515
|
Leong S, Teh BM, Kim AH. Characterization of otologic symptoms appearing after COVID-19 vaccination. Am J Otolaryngol 2023; 44:103725. [PMID: 36525812 PMCID: PMC9721195 DOI: 10.1016/j.amjoto.2022.103725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Anecdotal reports of sudden sensorineural hearing loss (SSNHL) following COVID-19 vaccination have emerged in the otolaryngology community. Studies have demonstrated no association between COVID-19 vaccination and SSNHL. We aim to characterize the spectrum of otologic symptoms following COVID-19 vaccination. METHODS A cross-sectional study of patients seen in the otology clinic at an academic center was performed. Patients completed a questionnaire on the development of new otologic symptoms within 4 weeks of COVID-19 vaccination. Diagnostic and audiometric data was collected retrospectively for patients reporting otologic symptoms. RESULTS Between May and July 2021, 500 patients were screened. Median age was 56.6 years old, with 59.4 % female and 40.2 % male. 420 patients (84.0 %) were vaccinated, with 58.4 % receiving Pfizer, 29.1 % receiving Moderna, and 3.8 % receiving Johnson & Johnson. 61 patients (14.5 %) reported one or more otologic symptoms within 4 weeks of vaccination, including 21 (5.0 %) with hearing loss, 26 (6.2 %) with tinnitus, 33 (7.9 %) with dizziness, and 19 (4.5 %) with vertigo. Of the 16 patients (3.2 %) reporting tinnitus with no associated hearing loss, 8 were diagnosed with subjective tinnitus and 4 were diagnosed with temporomandibular joint syndrome. Of the 18 patients reporting hearing loss, 11 had exacerbations of underlying pathologies (e.g. Meniere's disease, presbycusis) and 7 were newly diagnosed with SSNHL (1.4 %). CONCLUSIONS Patients reporting otologic symptoms following COVID-19 vaccination received various diagnoses of uncertain etiology. The incidence of SSNHL in these patients is comparable to the general otology patient population. Additional studies are required to determine the incidence of specific diagnoses following vaccination.
Collapse
Affiliation(s)
- Stephen Leong
- Columbia University Vagelos College of Physicians & Surgeons, New York, NY, United States of America
| | - Bing M Teh
- Department of Otolaryngology/Head and Neck Surgery, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, NY, United States of America; Department of Otolaryngology-Head & Neck Surgery, Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Ana H Kim
- Department of Otolaryngology/Head and Neck Surgery, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, NY, United States of America.
| |
Collapse
|
516
|
Park WB, Hwang YH, Cheong HJ. COVID-19 Vaccination in Korea. Infect Chemother 2023; 55:135-149. [PMID: 37021429 PMCID: PMC10079439 DOI: 10.3947/ic.2023.0023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Since December 2020, various coronavirus disease 2019 (COVID-19) vaccines have been developed and approved. As of February 2023, mRNA vaccines including bivalent vaccines (Pfizer/BioNTech, Moderna), recombinant protein vaccines (Novavax, SK Bioscience), and viral vector vaccines (AstraZeneca, Janssen) have been approved in Korea. COVID-19 vaccination can effectively reduce hospitalization and deaths due to symptomatic COVID-19, especially severe and critical COVID-19. The primary series vaccination against COVID-19 is recommended for all adults aged ≥18 years in Korea. Booster vaccination with the bivalent mRNA vaccine is available for those ≥12 years who have completed the primary series vaccination, regardless of the type of vaccine previously received, and is recommended for all adults. Booster vaccination can be administered since 90 days after the last dose. Localized and systemic adverse events following COVID-19 vaccination are relatively common and more frequently documented in younger age groups. Rare but potentially serious specialized adverse reactions include anaphylaxis, thrombosis with thrombocytopenia syndrome, myocarditis, and Guillain-Barré syndrome. Previous severe allergic reactions, such as anaphylaxis, to any COVID19 vaccine or vaccine component are considered a contraindication for vaccination. The indications and schedule for COVID-19 vaccination are subject to change based on further research results and the COVID-19 pandemic.
Collapse
Affiliation(s)
- Wan Beom Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Young Hoon Hwang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hee Jin Cheong
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea.
| |
Collapse
|
517
|
Ong MJY, Khoo CS, Lee YX, Poongkuntran V, Tang CK, Choong YJ, Hod R, Tan HJ. Safety and adverse events following COVID-19 vaccination among people with epilepsy: A cross-sectional study. Epilepsia Open 2023; 8:60-76. [PMID: 36214033 PMCID: PMC9874900 DOI: 10.1002/epi4.12658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/05/2022] [Indexed: 01/27/2023] Open
Abstract
OBJECTIVE Epilepsy is a non-communicable disease costing a massive burden globally. It is known that there is increased prevalence of morbidity and mortality following COVID-19 infection among people with epilepsy (PWE). However, there is limited information about the adverse events following COVID-19 immunization among PWE. Hence, this study aimed to assess the safety and adverse events following immunization (AEFI) of various COVID-19 vaccines among PWE from our centre, focusing on neurological AEFI. METHODS This cross-sectional study recruited 120 adult PWE from the Neurology Clinic of the Universiti Kebangsaan Malaysia Medical Centre (UKMMC). Consent-taking was conducted via synchronous or asynchronous approaches, followed by a phone call interview session. The interview collected socio-demographic information, epilepsy-related variables, and vaccination-related variables. Univariate analysis and multiple logistic regression analysis were done to confirm factors associated with the AEFI of COVID-19 vaccination. RESULTS Among all types of COVID-19 vaccines, most of the PWE received the Cominarty® COVID-19 vaccination (52.5%). Overall, local AEFI was the quickest to develop, with an average onset within a day. PWE with normal body mass index (BMI) had a higher risk of developing both local and systemic AEFI compared to those underweight and obese PWE (OR: 15.09, 95% CI 1.70-134.28, P = 0.02). SIGNIFICANCE COVID-19 vaccines are safe for PWE. AEFI among PWE are similar to those of the general population following COVID-19 vaccination. Therefore, clinicians should encourage PWE to take COVID-19 vaccines.
Collapse
Affiliation(s)
- Marjorie Jia Yi Ong
- Neurology Unit, Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia.,Department of Community Health, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Ching Soong Khoo
- Neurology Unit, Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Yi Xuan Lee
- Neurology Unit, Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Vaanee Poongkuntran
- Neurology Unit, Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Chia Khoi Tang
- Neurology Unit, Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Yu Joe Choong
- Neurology Unit, Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Rozita Hod
- Department of Community Health, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Hui Jan Tan
- Neurology Unit, Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
518
|
Vartanian K, Fish D, Gronowski B, Kenton N, Robicsek A. Patient-Reported Outcomes for Fully Vaccinated COVID-19 Patients Over 6 Weeks: The Experiences of Clinical Breakthrough Cases. THE PATIENT 2023; 16:105-116. [PMID: 36336751 PMCID: PMC9638265 DOI: 10.1007/s40271-022-00605-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 09/27/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND While coronavirus disease 2019 (COVID-19) vaccines have high rates of efficacy, fully vaccinated individuals can become infected with COVID-19. Among this population, symptoms tend to be less severe and shorter lasting. Less is known about how vaccinated individuals who contract COVID-19 experience the disease through patient-reported outcomes (PROs) and how this changes over time. OBJECTIVE The aim of this study was to describe the physical, mental, and social health PROs for fully vaccinated individuals who contracted COVID-19 over a 6-week period. DESIGN Prospective design using the Patient-Reported Outcomes Measurement Information System short-form (PROMIS-10) collected through a mobile application-based platform. PARTICIPANT 1114 fully vaccinated patients who tested positive for COVID-19 at a large US health system and engaged with the study on or after 1 March 2021 and reported onset of illness prior to 1 November 2021. MAIN MEASURES Global physical and mental health PROMIS-10 T-scores for the 6-week period, component PROMIS-10 questions for the 6-week period, and component PROMIS-10 questions restricted to a subset of participants for the first month to measure individual recovery were analyzed. KEY RESULTS Mean global physical and mental health T-scores increased over time and remained within one standard deviation of the population mean. At baseline, at least 40% of participants reported good health for all component questions except Fatigue (25%), and the proportion reporting good health increased over time for all questions, with the largest improvements in Fatigue (25.5 to 67.5%), Pain (59.1 to 82.8%), and Emotional Problems (42.3 to 62.5%). Over the first month, the greatest positive changes in individual recovery were observed for Fatigue (65.0%), Pain (53.0%), and Emotional Problems (41.1%); at least 30% of respondents reported no change in at least one category, and the greatest decreases were for Usual Social Activities (23.9%), Social Satisfaction (23.2%), and Mental Health (21.8%). CONCLUSIONS This study provides an important step towards better understanding the impact of 'breakthrough' COVID-19 infections on clinically engaged, fully vaccinated patients' physical and mental health to improve support for their treatment and recovery.
Collapse
Affiliation(s)
- Keri Vartanian
- Center for Outcomes Research and Education (CORE), Providence St. Joseph Health, 5251 NE Glisan Street, Portland, OR, USA
| | - Daniel Fish
- Center for Outcomes Research and Education (CORE), Providence St. Joseph Health, 5251 NE Glisan Street, Portland, OR, USA
| | - Benjamin Gronowski
- Center for Outcomes Research and Education (CORE), Providence St. Joseph Health, 5251 NE Glisan Street, Portland, OR, USA.
| | - Natalie Kenton
- Center for Outcomes Research and Education (CORE), Providence St. Joseph Health, 5251 NE Glisan Street, Portland, OR, USA
| | - Ari Robicsek
- Providence Research Network, 1801 Lind Ave SW, Renton, WA, USA
| |
Collapse
|
519
|
Oyebanji OA, Mylonakis E, Canaday DH. Vaccines for the Prevention of Coronavirus Disease 2019 in Older Adults. Infect Dis Clin North Am 2023; 37:27-45. [PMID: 36805013 PMCID: PMC9633624 DOI: 10.1016/j.idc.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Institutionalized and community-dwelling older adults have been greatly impacted by the coronavirus disease 2019 (COVID-19) pandemic with increased morbidity and mortality. The advent of vaccines and their widespread use in this population has brought about a dramatic turnaround in COVID-19 outcomes. The immunogenicity and effectiveness of the various vaccine options worldwide are discussed. Optimization of vaccine usage will still be important to maximize protection due to reduced initial immunity, development of variant strains, and fading of immunity over time. There are also lessons learned specific to older populations for future pandemics of novel pathogens.
Collapse
Affiliation(s)
- Oladayo A Oyebanji
- Case Western Reserve University, School of Medicine, 10900 Euclid Ave, BRB 1025, Cleveland, OH 44106-4984, USA
| | - Eleftherios Mylonakis
- Infectious Diseases Division, The Miriam Hospital and Rhode Island Hospital, Warren Alpert Medical School of Brown University, Rhode Island Hospital, 593 Eddy Street, POB, 3rd Floor, Suite 328/330, Providence, RI 02903, USA
| | - David H Canaday
- Case Western Reserve University, School of Medicine, 10900 Euclid Ave, BRB 1025, Cleveland, OH 44106-4984, USA; Geriatric Research, Education and Clinical Center, Cleveland Veterans Affairs Medical Center, 10900 Euclid Ave, BRB 1025, Cleveland, OH 44106-4984, USA.
| |
Collapse
|
520
|
Yan HY, Young YH. Vertigo/dizziness following COVID-19 vaccination. Am J Otolaryngol 2023; 44:103723. [PMID: 36502671 PMCID: PMC9721153 DOI: 10.1016/j.amjoto.2022.103723] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 11/28/2022] [Indexed: 12/07/2022]
Abstract
PURPOSE This study assessed the vertigo/dizziness in patients following COVID-19 vaccination. PATIENTS AND METHODS From July 2021 to June 2022, totaling 50 patients with dizzy spells following COVID-19 vaccination by AZ (AstraZeneca-Oxford University, AZD1222), BNT (Pfizer-BioNTech, BNT162b2) or Moderna (Moderna, mRNA-1273) vaccine were enrolled in this study. The interval from vaccination to the onset of vertigo/dizziness was compared with inter-episodic interval of vertigo/dizziness in the same patients, but without vaccination, during past one year (2020). RESULTS The incidences of severe systemic complication per 106 shots were 0.86 for Moderna vaccine, 1.22 for AZ vaccine, and 1.23 for BNT vaccine. Conversely, rate of post-vaccination vertigo/dizziness was noted in the Moderna group (66 %), followed by the AZ group (20 %) and the BNT (14 %) group, meaning that type of COVID-19 vaccine may affect various organ systems. The median time to the onset of vertigo/dizziness following vaccination is 10d, which is consistent with the onset of IgG production, and significantly less than inter-episodic interval (84d) in the same patients without vaccination. CONCLUSION Post-vaccination vertigo/dizziness can manifest as exacerbation of previous neurotological disorder. The median time to the onset of vertigo/dizziness following COVID-19 vaccination is 10d. Since the outcome is fair after supportive treatment, the immunomodulatory effect of the vaccines does not undermine the necessity of the COVID-19 vaccination.
Collapse
Affiliation(s)
- Hong-Yu Yan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Ho Young
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan; Deparment of Otolaryngology, Far Eastern Memorial Hospital, New Taipei, Taiwan.
| |
Collapse
|
521
|
Bioinformatics analysis based on high-throughput sequencing data to identify hub genes related to different clinical types of COVID-19. Funct Integr Genomics 2023; 23:71. [PMID: 36856850 PMCID: PMC9975444 DOI: 10.1007/s10142-023-00998-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023]
Abstract
This article aims to explore hub genes related to different clinical types of cases with COVID-19 and predict the therapeutic drugs related to severe cases. The expression profile of GSE166424 was divided into four data sets according to different clinical types of COVID-19 and then calculated the differential expression genes (DEGs). The specific genes of four clinical types of COVID-19 were obtained by Venn diagram and conducted enrichment analysis, protein-protein interaction (PPI) networks analysis, screening hub genes, and ROC curve analysis. The hub genes related to severe cases were verified in GSE171110, their RNA-specific expression tissues were obtained from the HPA database, and potential therapeutic drugs were predicted through the DGIdb database. There were 536, 266, 944, and 506 specific genes related to asymptomatic infections, mild, moderate, and severe cases, respectively. The hub genes of severe specific genes were AURKB, BRCA1, BUB1, CCNB1, CCNB2, CDC20, CDC6, KIF11, TOP2A, UBE2C, and RPL11, and also differentially expressed in GSE171110 (P < 0.05), and their AUC values were greater than 0.955. The RNA tissue specificity of AURKB, CDC6, KIF11, UBE2C, CCNB2, CDC20, TOP2A, BUB1, and CCNB1 specifically enhanced on lymphoid tissue; CCNB2, CDC20, TOP2A, and BUB1 specifically expressed on the testis. Finally, 55 drugs related to severe COVID-19 were obtained from the DGIdb database. Summary, AURKB, BRCA1, BUB1, CCNB1, CCNB2, CDC20, CDC6, KIF11, TOP2A, UBE2C, and RPL11 may be potential diagnostic biomarkers for severe COVID-19, which may affect immune and male reproductive systems. 55 drugs may be potential therapeutic drugs for severe COVID-19.
Collapse
|
522
|
Colangeli L, Aprile G, Carcone C, D'Adamo M, Medda E, Sbraccia P, Guglielmi V. Effects of COVID-19 vaccination on clinical outcomes in patients hospitalized in Internal Medicine during Omicron variant spreading. Intern Emerg Med 2023; 18:677-680. [PMID: 36637742 PMCID: PMC9837754 DOI: 10.1007/s11739-022-03185-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/13/2022] [Indexed: 01/14/2023]
Affiliation(s)
- Luca Colangeli
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
- Internal Medicine Unit and Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy.
| | - Gianna Aprile
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Clara Carcone
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Monica D'Adamo
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
- Internal Medicine Unit and Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Emanuela Medda
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore Sanità, Rome, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
- Internal Medicine Unit and Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Guglielmi
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
- Internal Medicine Unit and Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| |
Collapse
|
523
|
Paggi R, Barbiero A, Manciulli T, Miftode A, Tilli M, Lagi F, Mencarini J, Borchi B, Pozzi M, Bartalesi F, Spinicci M, Martini L, Coppola A, Nozzoli C, Peris A, Bonizzoli M, Pieralli F, Bartoloni A, Zammarchi L. Characteristics of COVID-19 vaccinated and unvaccinated patients admitted to Careggi University Hospital, Florence, Italy. Intern Emerg Med 2023; 18:821-830. [PMID: 36853393 PMCID: PMC9972322 DOI: 10.1007/s11739-023-03231-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 02/14/2023] [Indexed: 03/01/2023]
Abstract
More than 11.5 billion COVID-19 vaccine doses have been administered around the world. Although vaccine effectiveness for severe infections is reported to be 89.0%, breakthrough infections are common and may lead to severe outcome in fragile population. We conducted a real-world observational study on 420 COVID-19 admitted patients from July 2021 to January 2022 in a tertiary level Italian hospital. We collected patient's vaccination and SARS-CoV-2 serological status, SARS-CoV-2 treatments, oxygen supports, intensive (ICU) and subintensive (sub-ICU) care unit admissions, length of staying (LoS) and in-hospital mortality. One-hundred-seventy-two vaccinated and 248 unvaccinated patients were admitted during the study period. Vaccinated group (Vg) had a significantly more elevated Charlson Comorbidity Index than Unvaccinated group (UVg), and no statistical differences were found in terms of in-hospital mortality, LoS or ICU and sub-ICU admissions. Among Vg, anti-S antibodies were detected in 86.18% of patients (seropositives). Vaccinated seronegative patients' in-hospital mortality was significantly higher than vaccinated seropositive patients (33.33% vs 10.69%, p = 0.0055): in particular, mortality rate in 45-69 years old population was higher in vaccinated seronegative group, and comparable in patients ≥ 70 years old. No differences in terms of outcome were registered between Vg and UVg, taking into account that Vg was considerably older and with more comorbidities. In line with other recent observations, higher mortality rate was evidenced for seronegative vaccinated patients. Primary prophylaxis and early treatments result to be necessary, especially for older and immunosuppressed populations.
Collapse
Affiliation(s)
- Riccardo Paggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Anna Barbiero
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Tommaso Manciulli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Andreea Miftode
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Marta Tilli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Filippo Lagi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Jessica Mencarini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Beatrice Borchi
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Marco Pozzi
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Filippo Bartalesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Michele Spinicci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Martini
- Internal Medicine Unit 2, Careggi University Hospital, Florence, Italy
| | | | - Carlo Nozzoli
- Internal Medicine Unit 1, Careggi University Hospital, Florence, Italy
| | - Adriano Peris
- Intensive Care Unit and Regional ECMO Referral Centre, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Manuela Bonizzoli
- Intensive Care Unit and Regional ECMO Referral Centre, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Filippo Pieralli
- High-Intensity Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Zammarchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
524
|
Zerbo O, Ray GT, Fireman B, Layefsky E, Goddard K, Lewis E, Ross P, Omer S, Greenberg M, Klein NP. Maternal SARS-CoV-2 vaccination and infant protection against SARS-CoV-2 during the first six months of life. Nat Commun 2023; 14:894. [PMID: 36854660 PMCID: PMC9974935 DOI: 10.1038/s41467-023-36547-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/07/2023] [Indexed: 03/02/2023] Open
Abstract
We examined the effectiveness of maternal vaccination against SARS-CoV-2 infection in 30,311 infants born at Kaiser Permanente Northern California from December 15, 2020, to May 31, 2022. Using Cox regression, the effectiveness of ≥2 doses of COVID-19 vaccine received during pregnancy was 84% (95% confidence interval [CI]: 66, 93), 62% (CI: 39, 77) and 56% (CI: 34,71) during months 0-2, 0-4 and 0- 6 of a child's life, respectively, in the Delta variant period. In the Omicron variant period, the effectiveness of maternal vaccination in these three age intervals was 21% (CI: -21,48), 14% (CI: -9,32) and 13% (CI: -3,26), respectively. Over the entire study period, the incidence of hospitalization for COVID-19 was lower during the first 6 months of life among infants of vaccinated mothers compared with infants of unvaccinated mothers (21/100,000 person-years vs. 100/100,000 person-years). Maternal vaccination was protective, but protection was lower during Omicron than during Delta. Protection during both periods decreased as infants aged.
Collapse
Affiliation(s)
- Ousseny Zerbo
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA, USA.
| | - G Thomas Ray
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA, USA
| | - Bruce Fireman
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA, USA
| | - Evan Layefsky
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA, USA
| | - Kristin Goddard
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA, USA
| | - Edwin Lewis
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA, USA
| | - Pat Ross
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA, USA
| | - Saad Omer
- Yale University, Institute for Global Health, New Haven, CT, USA
- Department of Internal Medicine (Infectious Diseases), Yale School of Medicine, New Haven, CT, USA
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Mara Greenberg
- Obstetrics and Gynecology, Kaiser Permanente Northern California Oakland, Oakland, CA, USA
- Regional Perinatal Service Center, Kaiser Permanente Northern California, Santa Clara, CA, USA
| | - Nicola P Klein
- Kaiser Permanente Northern California, Vaccine Study Center, Oakland, CA, USA
| |
Collapse
|
525
|
Efficacy of SARS-CoV-2 vaccines and the dose-response relationship with three major antibodies: a systematic review and meta-analysis of randomised controlled trials. THE LANCET. MICROBE 2023; 4:e236-e246. [PMID: 36868258 PMCID: PMC9974155 DOI: 10.1016/s2666-5247(22)00390-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 03/04/2023]
Abstract
BACKGROUND The efficacy of SARS-CoV-2 vaccines in preventing severe COVID-19 illness and death is uncertain due to the rarity of data in individual trials. How well the antibody concentrations can predict the efficacy is also uncertain. We aimed to assess the efficacy of these vaccines in preventing SARS-CoV-2 infections of different severities and the dose-response relationship between the antibody concentrations and efficacy. METHODS We did a systematic review and meta-analysis of randomised controlled trials (RCTs). We searched PubMed, Embase, Scopus, Web of Science, Cochrane Library, WHO, bioRxiv, and medRxiv for papers published between Jan 1, 2020 and Sep 12, 2022. RCTs on the efficacy of SARS-CoV-2 vaccines were eligible. Risk of bias was assessed using the Cochrane tool. A frequentist, random-effects model was used to combine efficacy for common outcomes (ie, symptomatic and asymptomatic infections) and a Bayesian random-effects model was used for rare outcomes (ie, hospital admission, severe infection, and death). Potential sources of heterogeneity were investigated. The dose-response relationships of neutralising, spike-specific IgG and receptor binding domain-specific IgG antibody titres with efficacy in preventing SARS-CoV-2 symptomatic and severe infections were examined by meta-regression. This systematic review is registered with PROSPERO, CRD42021287238. FINDINGS 28 RCTs (n=286 915 in vaccination groups and n=233 236 in placebo groups; median follow-up 1-6 months after last vaccination) across 32 publications were included in this review. The combined efficacy of full vaccination was 44·5% (95% CI 27·8-57·4) for preventing asymptomatic infections, 76·5% (69·8-81·7) for preventing symptomatic infections, 95·4% (95% credible interval 88·0-98·7) for preventing hospitalisation, 90·8% (85·5-95·1) for preventing severe infection, and 85·8% (68·7-94·6) for preventing death. There was heterogeneity in the efficacy of SARS-CoV-2 vaccines against asymptomatic and symptomatic infections but insufficient evidence to suggest whether the efficacy could differ according to the type of vaccine, age of the vaccinated individual, and between-dose interval (p>0·05 for all). Vaccine efficacy against symptomatic infection waned over time after full vaccination, with an average decrease of 13·6% (95% CI 5·5-22·3; p=0·0007) per month but can be enhanced by a booster. We found a significant non-linear relationship between each type of antibody and efficacy against symptomatic and severe infections (p<0·0001 for all), but there remained considerable heterogeneity in the efficacy, which cannot be explained by antibody concentrations. The risk of bias was low in most studies. INTERPRETATION The efficacy of SARS-CoV-2 vaccines is higher for preventing severe infection and death than for preventing milder infection. Vaccine efficacy wanes over time but can be enhanced by a booster. Higher antibody titres are associated with higher estimates of efficacy but precise predictions are difficult due to large unexplained heterogeneity. These findings provide an important knowledge base for interpretation and application of future studies on these issues. FUNDING Shenzhen Science and Technology Programs.
Collapse
|
526
|
Konje JC, Al Beloushi M, Ahmed B. Immunisation against COVID-19 in Pregnancy and of Women Planning Pregnancy. Viruses 2023; 15:v15030621. [PMID: 36992330 PMCID: PMC10059008 DOI: 10.3390/v15030621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 03/03/2023] Open
Abstract
Following reports of the first human SARS-CoV2 infection in December 2019 from Wuhan Province, China, there was such rapid spread that by March 2021, the World Health Organization (WHO) had declared a pandemic. Over 6.5 million people have died from this infection worldwide, although this is most likely an underestimate. Until vaccines became available, mortality and severe morbidity were costly in terms of life lost as well as the cost of supporting the severely and acutely ill. Vaccination changed the landscape, and following worldwide adoption, life has gradually been returning to normal. The speed of production of the vaccines was unprecedented and undoubtedly ushered in a new era in the science of fighting infections. The developed vaccines were on the already known platforms for vaccine delivery: inactivated virus, virus vector, virus-like particles (VLP) subunit, DNA and mRNA. The mRNA platform was used for the first time to deliver vaccines to humans. An understanding of these platforms and the pros and cons of each are important for clinicians who are often challenged by the recipients on the advantages and risks of these vaccines. These vaccines have so far and reassuringly been shown to be safe in reproduction (with no effect on gametes) and pregnancy (not associated with congenital malformations). However, safety remains paramount and continuing vigilance is critical, especially against rare fatal complications such as vaccine-induced thrombocytopenia and myocarditis. Finally, the waning immunity months after vaccination means repeated immunisation is likely to be ongoing, but just how often and how many such revaccinations should be recommended remains uncertain. Research into other vaccines and alternate delivery methods should continue as this infection is likely to be around for a long time.
Collapse
Affiliation(s)
- Justin C. Konje
- Feto-Maternal Centre Al Markhiya, Doha P.O. Box 34181, Qatar
- Obstetrics and Gynecology Department, Weill Cornell Medicine Qatar, Doha P.O. Box 24144, Qatar
- Obstetrics and Gynaecology, Department of Health Sciences, University of Leicester, Leicester LE2 7LX, UK
- Correspondence: ; Tel.: +974-7777-8375
| | - Mariam Al Beloushi
- Women’s Wellness and Research Centre, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar
| | - Badreldeen Ahmed
- Feto-Maternal Centre Al Markhiya, Doha P.O. Box 34181, Qatar
- Obstetrics and Gynecology Department, Weill Cornell Medicine Qatar, Doha P.O. Box 24144, Qatar
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
527
|
Tsuchiya Y, Tamura H, Fujii K, Numaguchi H, Toyoizumi K, Liu T, Le Gars M, Cárdenas V, Eto T. Safety, reactogenicity, and immunogenicity of Ad26.COV2.S: Results of a phase 1, randomized, double-blind, placebo-controlled COVID-19 vaccine trial in Japan. Vaccine 2023; 41:1602-1610. [PMID: 36732164 PMCID: PMC9812825 DOI: 10.1016/j.vaccine.2023.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/10/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
BACKGROUND This study evaluated safety, reactogenicity, and immunogenicity of a 2-month homologous booster regimen of Ad26.COV2.S in Japanese adults. METHODS In this multicenter, placebo-controlled, Phase 1 trial, adults (Cohort 1, aged 20-55 years, N = 125; Cohort 2, aged ≥ 65 years, N = 125) were randomized 2:2:1 to receive Ad26.COV2.S 5 × 1010 viral particles (vp), Ad26.COV2.S 1 × 1011 vp, or placebo, followed by a homologous booster 56 days later. Safety, reactogenicity, and immunogenicity were assessed. RESULTS Two hundred participants received Ad26.COV2.S and 50 received placebo. The most frequent solicited local adverse event (AE) was vaccination-site pain, and the most frequent solicited systemic AEs were fatigue, myalgia, and headache. After primary vaccination, neutralizing and binding antibody levels increased through Day 57 (post-prime) in both cohorts. Fourteen days after boosting (Day 71), neutralizing antibody geometric mean titers (GMTs) had almost reached their peak value in Cohort 1 (5 × 1010 vp: GMT = 1049; 1 × 1011 vp: GMT = 1470) and peaked in Cohort 2 (504; 651); at Day 85, GMTs had declined minimally in Cohort 2. For both cohorts, binding antibody levels peaked at Day 71 with minimal decline at Day 85. CONCLUSION A single dose and homologous Ad26.COV2.S booster increased antibody responses with an acceptable safety profile in Japanese adults (ClinicalTrials.gov Identifier: NCT04509947).
Collapse
Affiliation(s)
- Yumi Tsuchiya
- Research and Development, Janssen Pharmaceutical K.K., Tokyo, Japan.
| | - Hiroshi Tamura
- Research and Development, Janssen Pharmaceutical K.K., Tokyo, Japan
| | - Koji Fujii
- Research and Development, Janssen Pharmaceutical K.K., Tokyo, Japan
| | | | - Kiichiro Toyoizumi
- Statistics and Decision Sciences, Janssen Pharmaceutical K.K., Tokyo, Japan
| | - Tina Liu
- Clinical and Statistical Programming, Janssen China Research and Development, Beijing, China
| | | | | | - Takashi Eto
- Souseikai Hakata Clinic, Fukuoka-city, Fukuoka, Japan
| |
Collapse
|
528
|
The COVID-19 Vaccination Coverage in ICU Patients with Severe COVID-19 Infection in a Country with Low Vaccination Coverage-A National Retrospective Analysis. J Clin Med 2023; 12:jcm12051749. [PMID: 36902535 PMCID: PMC10003614 DOI: 10.3390/jcm12051749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Romania is one of the European countries with low COVID-19 vaccination coverage. The main goal of this study was to describe the COVID-19 vaccination status in patients admitted to Romanian ICUs with a severe COVID-19 infection. The study describes the patients' characteristics according to their vaccination status and evaluates the association between vaccination status and ICU mortality. METHODS This retrospective, observational, multicenter study included patients with confirmed vaccination status admitted to Romanian ICUs from January 2021 to March 2022. RESULTS Two thousand, two hundred and twenty-two patients with confirmed vaccination status were included. Five point one three percent of patients were vaccinated with two vaccine doses and one point seventeen percent of patients were vaccinated with one vaccine dose. The vaccinated patients showed a higher rate of comorbidities but had similar clinical characteristics at ICU admission and lower mortality rates compared to non-vaccinated patients. Vaccinated status and higher Glasgow Coma Scale at ICU admission were independently associated with ICU survival. Ischemic heart disease, chronic kidney disease, higher SOFA score at ICU admission and the need for mechanical ventilation in ICU were independently associated with ICU mortality. CONCLUSION Lower rates of ICU admission were observed in fully vaccinated patients even in a country with low vaccination coverage. The ICU mortality was lower for fully vaccinated patients compared to non-vaccinated patients. The benefit of vaccination on ICU survival could be more important in patients with associated comorbidities.
Collapse
|
529
|
Dhamanti I, Suwantika AA, Adlia A, Yamani LN, Yakub F. Adverse Reactions of COVID-19 Vaccines: A Scoping Review of Observational Studies. Int J Gen Med 2023; 16:609-618. [PMID: 36845341 PMCID: PMC9951602 DOI: 10.2147/ijgm.s400458] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
The COVID-19 pandemic had a severe global impact. A range of campaigns and activities, including vaccines, are being implemented to counteract this pandemic. Using observational data, the goal of this scoping review is to identify adverse events connected with COVID-19 vaccinations. We conduct a scoping study and searched three databases from the start of the COVID-19 pandemic in 2020 through June 2022. Based on our criteria and searched keywords, the review included eleven papers in total, with the majority of the studies being conducted in developed countries. The study populations varied and included general community populations, healthcare professionals, military forces, and patients with systemic lupus and cancer. This study includes vaccines from Pfizer-BioNTech, Oxford-AstraZeneca, Sinopharm, and Moderna. The COVID-19 vaccine-related adverse events were classified into three types: local side effects, systemic side effects, and other side effects such as allergies. The adverse reactions to COVID-19 vaccines are mild to moderate in severity, with no significant influence or interference in individual daily activities and no unique patterns in cause of death among vaccine-related deaths. According to the findings of these investigations, the COVID-19 vaccine is safe to administer and induces protection. It is vital to convey accurate information to the public about vaccination side effects, potential adverse responses, and the safety level of the vaccines supplied. Multiple strategies must be implemented at the individual, organizational, and population levels to eliminate vaccine hesitance. Future studies could investigate the vaccine's effect on people of various ages and medical conditions.
Collapse
Affiliation(s)
- Inge Dhamanti
- Department of Health Policy and Administration, Faculty of Public Health, Universitas Airlangga, Surabaya, Indonesia
- Center for Patient Safety Research, Universitas Airlangga, Surabaya, Indonesia
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Auliya A Suwantika
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
- Center for Health Technology Assessment, Universitas Padjadjaran, Bandung, Indonesia
| | - Amirah Adlia
- Department of Pharmaceutics, School of Pharmacy, Institut Teknologi Bandung, Bandung, Indonesia
| | - Laura Navika Yamani
- Division Epidemiology, Faculty of Public Health, Universitas Airlangga, Surabaya, Indonesia
- Research Center on Global Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Fitri Yakub
- Malaysia-Japan International Institute of Technology, Universiti Teknologi Malaysia, Skudai, Malaysia
| |
Collapse
|
530
|
Xu Z, Wei D, Zhang H, Demongeot J. A Novel Mathematical Model That Predicts the Protection Time of SARS-CoV-2 Antibodies. Viruses 2023; 15:v15020586. [PMID: 36851801 PMCID: PMC9962246 DOI: 10.3390/v15020586] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/10/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Infectious diseases such as SARS-CoV-2 pose a considerable threat to public health. Constructing a reliable mathematical model helps us quantitatively explain the kinetic characteristics of antibody-virus interactions. A novel and robust model is developed to integrate antibody dynamics with virus dynamics based on a comprehensive understanding of immunology principles. This model explicitly formulizes the pernicious effect of the antibody, together with a positive feedback stimulation of the virus-antibody complex on the antibody regeneration. Besides providing quantitative insights into antibody and virus dynamics, it demonstrates good adaptivity in recapturing the virus-antibody interaction. It is proposed that the environmental antigenic substances help maintain the memory cell level and the corresponding neutralizing antibodies secreted by those memory cells. A broader application is also visualized in predicting the antibody protection time caused by a natural infection. Suitable binding antibodies and the presence of massive environmental antigenic substances would prolong the protection time against breakthrough infection. The model also displays excellent fitness and provides good explanations for antibody selection, antibody interference, and self-reinfection. It helps elucidate how our immune system efficiently develops neutralizing antibodies with good binding kinetics. It provides a reasonable explanation for the lower SARS-CoV-2 mortality in the population that was vaccinated with other vaccines. It is inferred that the best strategy for prolonging the vaccine protection time is not repeated inoculation but a directed induction of fast-binding antibodies. Eventually, this model will inform the future construction of an optimal mathematical model and help us fight against those infectious diseases.
Collapse
Affiliation(s)
- Zhaobin Xu
- Department of Life Science, Dezhou University, Dezhou 253023, China
- Correspondence: (Z.X.); (J.D.)
| | - Dongqing Wei
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Hongmei Zhang
- Department of Life Science, Dezhou University, Dezhou 253023, China
| | - Jacques Demongeot
- Laboratory AGEIS EA 7407, Team Tools for e-Gnosis Medical, Faculty of Medicine, University Grenoble Alpes (UGA), 38700 La Tronche, France
- Correspondence: (Z.X.); (J.D.)
| |
Collapse
|
531
|
Renal Biopsy Diagnosis of Acute Tubular Injury after Pfizer-BioNTech COVID-19 Vaccination: A Case Report. Vaccines (Basel) 2023; 11:vaccines11020464. [PMID: 36851341 PMCID: PMC9965903 DOI: 10.3390/vaccines11020464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a severe respiratory infection that can be fatal in unvaccinated individuals; however, acute kidney injury (AKI) is a rare adverse reaction to COVID-19 vaccination. AKI resulting from multiple conditions can have severe consequences, including end-stage renal failure, if not treated with immunosuppressive agents. However, acute tubular injury (ATI) as the sole cause of AKI has not been previously reported. Herein, we discuss an obese 54-year-old man with type 2 diabetes who received four COVID-19 vaccines; three from Pfizer and one from Moderna. Diabetic retinopathy, urinary protein, and occult blood were absent with no other underlying diseases. There was no history of COVID-19 infection. He was referred to our hospital 5 days after receiving the fourth Pfizer-BioNTech COVID-19 vaccine dose with stage 3 AKI. Urinary findings revealed new proteinuria and glomerular occult blood. Physical examination and infection testing were unremarkable. Steroids were introduced on admission for rapidly progressive glomerulonephritis. A renal biopsy performed on Day 2 revealed only ATI. Therefore, steroids were discontinued on Day 5, after which renal function recovered spontaneously, and urinalysis abnormalities disappeared. Renal function remained normal during follow-up. We report a case of AKI with severe renal dysfunction after COVID-19 vaccination, wherein renal biopsy effectively determined the disease status (ATI), which did not require immunosuppressive treatment.
Collapse
|
532
|
Song Z, Luo Q, Wan L, Zhu Q, Liu R, Yin X, Lu X, Wei L, Xiang Z, Zou Y. Analysis of Antibodies Induced after SARS-CoV-2 Vaccination Using Antigen Coded Bead Array Luminex Technology. Vaccines (Basel) 2023; 11:442. [PMID: 36851319 PMCID: PMC9964277 DOI: 10.3390/vaccines11020442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/02/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
Objectives. Since the outbreak of SARS-CoV-2 in late 2019, nearly 12.2 billion doses of the COVID-19 vaccine have been administered worldwide; however, the humoral immune responses induced by different types of vaccines are yet to be fully validated. Methods. We analyzed antibody levels in 100 serum samples after vaccination with different types of COVID-19 vaccines and their reactivity against the RBD antigen of Delta and Omicron variants using a bead-based microarray. Results. Elevated levels of anti-wild-type (WT)-RBD IgG and anti-WT-NP IgG were detected in participants who received two doses of the inactivated vaccines (CoronaVac or BBIBP-CorV) and three doses of the recombinant spike protein vaccine (ZF2001), indicating that antibody responses to SARS-CoV-2 were generated regardless of the vaccine administered. We found highly correlated levels of serum anti-RBD IgG and anti-NP IgG (r = 0.432, p < 0.001). We observed that the antibodies produced in vivo after COVID-19 vaccination still reacted with variants of SARS-CoV-2 (p < 0.0001). Conclusions. Our results show that high levels of specific antibodies can be produced after completion of COVID-19 vaccination (two doses of the inactivated vaccines or three doses of ZF2001), with some degree of cross-reactivity to the RBD antigen of Delta and Omicron variants, and provide an accessible and practical experimental method for post-vaccination antibody detection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yizhou Zou
- Department of Immunology, School of Basic Medical of Central South University, Changsha 410083, China
| |
Collapse
|
533
|
Oliveira I, Ferreira I, Jacob B, Cardenas K, Cerni F, Baia-da-Silva D, Arantes E, Monteiro W, Pucca M. Harnessing the Power of Venomous Animal-Derived Toxins against COVID-19. Toxins (Basel) 2023; 15:159. [PMID: 36828473 PMCID: PMC9967918 DOI: 10.3390/toxins15020159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/11/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Animal-derived venoms are complex mixtures of toxins triggering important biological effects during envenomings. Although venom-derived toxins are known for their potential of causing harm to victims, toxins can also act as pharmacological agents. During the COVID-19 pandemic, there was observed an increase in in-depth studies on antiviral agents, and since, to date, there has been no completely effective drug against the global disease. This review explores the crosstalk of animal toxins and COVID-19, aiming to map potential therapeutic agents derived from venoms (e.g., bees, snakes, scorpions, etc.) targeting COVID-19.
Collapse
Affiliation(s)
- Isadora Oliveira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Isabela Ferreira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Beatriz Jacob
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Kiara Cardenas
- Medical School, Federal University of Roraima, Boa Vista 69310-000, RR, Brazil
| | - Felipe Cerni
- Health Sciences Postgraduate Program, Federal University of Roraima, Boa Vista 69310-000, RR, Brazil
| | - Djane Baia-da-Silva
- Institute of Clinical Research Carlos Borborema, Dr. Heitor Vieira Dourado Tropical Medicine Foundation, Manaus 69850-000, AM, Brazil
- Postgraduate Program in Tropical Medicine, School of Health Sciences, Amazonas State University, Manaus 69850-000, AM, Brazil
- Department of Collective Health, Faculty of Medicine, Federal University of Amazonas, Manaus 69077-000, AM, Brazil
- Leônidas and Maria Deane Institute, Fiocruz Amazônia, Manaus 69057-070, AM, Brazil
- Faculty of Pharmacy, Nilton Lins University, Manaus 69058-040, AM, Brazil
| | - Eliane Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Wuelton Monteiro
- Institute of Clinical Research Carlos Borborema, Dr. Heitor Vieira Dourado Tropical Medicine Foundation, Manaus 69850-000, AM, Brazil
- Postgraduate Program in Tropical Medicine, School of Health Sciences, Amazonas State University, Manaus 69850-000, AM, Brazil
| | - Manuela Pucca
- Medical School, Federal University of Roraima, Boa Vista 69310-000, RR, Brazil
- Health Sciences Postgraduate Program, Federal University of Roraima, Boa Vista 69310-000, RR, Brazil
- Postgraduate Program in Tropical Medicine, School of Health Sciences, Amazonas State University, Manaus 69850-000, AM, Brazil
| |
Collapse
|
534
|
Kim S, Seok HY. Evaluation of the safety profile of COVID-19 vaccines in patients with MS, NMOSD, and MOGAD. Neurol Sci 2023; 44:1841-1848. [PMID: 36781562 PMCID: PMC9924883 DOI: 10.1007/s10072-023-06676-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 02/08/2023] [Indexed: 02/15/2023]
Abstract
INTRODUCTION Vaccination against the coronavirus disease 2019 (COVID-19) is recommended for patients with multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), and myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD). However, vaccine safety in these patients taking immunotherapeutic agents is unclear as they were not included in the vaccine trials. OBJECTIVES To evaluate the safety of COVID-19 vaccines in patients with MS, NMOSD, and MOGAD. METHODS We reviewed the medical records of MS, NMOSD, and MOGAD patients at the Keimyung University Dongsan Hospital. Information regarding vaccination schedules and adverse events was collected. RESULTS A total of 56 patients (19, 22, and 15 patients with MS, NMOSD, and MOGAD, respectively) with a median age of 48.18 ± 15.72 years (range, 16-81 years) were included. Of them, 42 (75.0%) were female. In total, 76.8% (43/56) of all patients were vaccinated, and the vaccination rate was the highest for NMOSD patients (81.8%) and the lowest for MS patients (68.4%). All vaccinated patients were administered mRNA vaccines at least once in single or multiple vaccination doses. Only 3 of 43 (7.0%) vaccinated patients experienced clinical relapse following vaccination. Facial sensory changes with a brainstem lesion developed in an MS patient taking dimethyl fumarate, while myelitis occurred in a MOGAD patient receiving azathioprine maintenance therapy. The first episode of optic neuritis occurred in a patient who was later diagnosed with MOGAD. CONCLUSIONS Our study demonstrated a favorable safety profile with no serious adverse events associated with COVID-19 vaccines in patients with MS, NMOSD, and MOGAD.
Collapse
Affiliation(s)
- Sohyeon Kim
- Department of Neurology, Dongsan Hospital, Keimyung University School of Medicine, 1035 Dalgubeol-daero, Dalseo-gu, Daegu, 42601 Republic of Korea
| | - Hung Youl Seok
- Department of Neurology, Dongsan Hospital, Keimyung University School of Medicine, 1035 Dalgubeol-daero, Dalseo-gu, Daegu, 42601, Republic of Korea. .,Department of Neurology, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
535
|
Claaß LV, Mayr P, Paschold L, Weber T, Terziev D, Jehs B, Brill R, Dober J, Märkl B, Wickenhauser C, Czapiewski P, Trepel M, Claus R, Binder M. No association of malignant B-cell non-Hodgkin lymphomas with ipsilateral SARS-CoV-2 vaccination. Cancer Med 2023; 12:9313-9321. [PMID: 36775947 PMCID: PMC10166887 DOI: 10.1002/cam4.5687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/10/2023] [Accepted: 02/01/2023] [Indexed: 02/14/2023] Open
Abstract
PURPOSE SARS-CoV-2 vaccines cause acute ipsilateral lymph node swelling in an important proportion of vaccines. Thus far, no malignant lymphadenopathies have been reported in temporal context to vaccination in the ipsilateral draining lymph node areas. EXPERIMENTAL DESIGN Prompted by two cases with unilateral axillary lymphomas that occurred ipsilaterally to prior SARS-CoV-2 vaccination, we systematically retrieved all B-cell non-Hodgkin lymphomas at two German University Medical Centers diagnosed before and after introduction of SARS-CoV-2 vaccines in Germany. Available lymphoma tissue (n=19) was subjected to next-generation immunosequencing of the IGH locus. Malignant clonotypes were mined in the CoVabDab database and published data sets from 342 uninfected individuals, 55 individuals 28 days after anti-SARS-CoV-2 vaccination and 139 individuals with acute COVID-19 together encompassing over 1 million CDR3 sequences in total. RESULTS Of 313 newly diagnosed cases in the two centers and observation periods, 27 unilateral manifestations in the defined deltoid draining regions were identified. The majority thereof were diffuse large B-cell lymphomas (18 of 27 cases). Eleven unilateral cases were diagnosed in the era of SARS-CoV-2 vaccination and 16 in the control period before introduction of such vaccines. Of the 11 unilateral lymphomas that occurred during the vaccination period, ten had received a SARS-CoV-2 vaccine prior to lymphoma diagnosis. These cases were further evaluated. While left-sided were more frequent than right-sided lymphomas (19 vs 8 cases), no statistically significant association of vaccination site and laterality of the lymphoma manifestation was found. The unilateral lymphomas showed a normal range of B-cell receptors typically found in these lymphoma subtypes with no evidence for anti-SARS-CoV-2 sequences in the malignant clonotype. CONCLUSIONS Together, we found no evidence that the current SARS-CoV-2 vaccines could serve as a trigger for lymphomagenesis in the draining lymph node areas of the deltoid region used for vaccination.
Collapse
Affiliation(s)
- Luise Victoria Claaß
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Patrick Mayr
- Department of Hematology and Oncology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Lisa Paschold
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Thomas Weber
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Denis Terziev
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Bertram Jehs
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Richard Brill
- Clinic and Policlinic of Radiology, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Johannes Dober
- Clinic and Policlinic of Radiology, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Bruno Märkl
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, University Hospital Halle (Saale), Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Piotr Czapiewski
- Department of Pathology, Medical Faculty, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany.,Department of Pathology Dessau Medical Centre, Institute of Pathology, Dessau, Germany
| | - Martin Trepel
- Department of Hematology and Oncology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Rainer Claus
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Augsburg, Germany.,Comprehensive Cancer Center Augsburg (CCCA), Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Mascha Binder
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
536
|
Vosko I, Zirlik A, Bugger H. Impact of COVID-19 on Cardiovascular Disease. Viruses 2023; 15:508. [PMID: 36851722 PMCID: PMC9962056 DOI: 10.3390/v15020508] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a viral infection with the novel severe acute respiratory distress syndrome corona virus 2 (SARS-CoV-2). Until now, more than 670 million people have suffered from COVID-19 worldwide, and roughly 7 million death cases were attributed to COVID-19. Recent evidence suggests an interplay between COVID-19 and cardiovascular disease (CVD). COVID-19 may serve as a yet underappreciated CVD risk modifier, including risk factors such as diabetes mellitus or arterial hypertension. In addition, recent data suggest that previous COVID-19 may increase the risk for many entities of CVD to an extent similarly observed for traditional cardiovascular (CV) risk factors. Furthermore, increased CVD incidence and worse clinical outcomes in individuals with preexisting CVD have been observed for myocarditis, acute coronary syndrome, heart failure (HF), thromboembolic complications, and arrhythmias. Direct and indirect mechanisms have been proposed by which COVID-19 may impact CVD and CV risk, including viral entry into CV tissue or by the induction of a massive systemic inflammatory response. In the current review, we provide an overview of the literature reporting an interaction between COVID-19 and CVD, review potential mechanisms underlying this interaction, and discuss preventive and treatment strategies and their interference with CVD that were evaluated since the onset of the COVID-19 pandemic.
Collapse
Affiliation(s)
| | | | - Heiko Bugger
- Department of Cardiology, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
537
|
Guo Y, Cao XS, Yang HT, Zhou MG, Yu B. Global incidence pattern and factors associated with common cutaneous reactions related to COVID-19 vaccination of 2.55 million participants in real-world settings: A systematic review and meta-analysis. J Glob Health 2023; 13:06008. [PMID: 36757823 PMCID: PMC9910561 DOI: 10.7189/jogh.13.06008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
Background Understanding the incidence pattern of cutaneous reactions is crucial for promoting COVID-19 vaccination. We aimed to report the global incidence pattern of, and factors associated with common cutaneous reactions related to COVID-19 vaccination in real-world settings. Methods We searched five databases (PubMed, Web of Science, Embase, CNKI, and Wanfang) from inception to May 13, 2022, for studies reporting the incidence of common cutaneous reactions related to COVID-19 vaccines in real-world settings. The outcomes were the systematic skin reactions (rash and urticaria) and the local injection site reactions (pain, swelling, redness, and erythema). We conducted random-effects meta-analyses and explored associated factors using multi-step statistical analyses. Results We included 35 studies and assessed 2 549 968 participants from 23 countries. The pooled incidence of overall systemic skin reactions was 3.8% (95% confidence interval (CI) = 2.4%-5.5%) with short duration (about one week). Specifically, the pooled incidence rates of rash and urticaria were 3.0% (95% CI = 2.1%-3.9%) and 1.1% (95% CI = 0.7%-1.5%), respectively. For overall local injection site reactions, the pooled incidence was 72.4% (95% CI = 65.7%-78.7%) with short duration (1 to 4.5 days). Except for local pain (72.2%, 95% CI = 65.3%-78.5%), other localized reactions had low incidence, including swelling (13.3%, 95% CI = 9.5%-17.7%), redness (11.5%, 95% CI = 5.7%-19.0%), and erythema (5.8%, 95% CI = 0.7%-15.4%). Geographically, different distribution patterns were observed for these reactions. Regarding associated factors, mRNA vaccines showed lower incidence of urticaria (P < 0.001). Asia population showed higher incidence of urticaria (P < 0.001). We observed lower incidence rates of overall local injection site reactions and pain among inactivated vaccines (P < 0.001). We found no significant difference among reactions between the first and the second dose of vaccines. Conclusions We examined the global incidence pattern of common cutaneous reactions related to COVID-19 vaccination and found low incidence and short duration of systemic skin reactions and local injection site reactions (except for pain); discrepancies in these reactions were observed across different vaccine types. The cutaneous side effects related to COVID-19 vaccination do not seem to cause concern. Registration PROSPERO: CRD42021258012.
Collapse
Affiliation(s)
- Yang Guo
- Department of Dermatology, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Xue-Shan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Hua-Tong Yang
- Department of Statistics and Data Science, Southern University of Science and Technology, Shenzhen, China,Department of Statistics, University of California Berkeley, Berkeley, California, USA
| | - Meng-Ge Zhou
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Bo Yu
- Department of Dermatology, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| |
Collapse
|
538
|
Darby B, Alexander V, Murphy J. SARS-CoV-2 vaccine breakthrough infections in Virginia, January 17, 2021 - June 30, 2021. Vaccine 2023; 41:1295-1298. [PMID: 36690560 PMCID: PMC9852316 DOI: 10.1016/j.vaccine.2023.01.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/04/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
As COVID-19 vaccines moved from the controlled environment of clinical trials to use in real-world settings, it has been important to evaluate vaccine effectiveness. A retrospective cohort study was designed to identify cases of SARS-CoV-2 infection that occurred between January 17-June 30, 2021 in fully vaccinated Virginia residents. Of the fully vaccinated population of Virginia at the end of the study period (N = 4,271,505), 2445 (0.057 %) were reported to have experienced a vaccine breakthrough infection. Of those, 183 (7.5 %) were reported to have been hospitalized for COVID-19 and 53 (2.2 %) died from COVID-19. There were significant differences in vaccine effectiveness over time between both mRNA vaccines and the Janssen vaccine. Increasing age, pre-existing medical conditions, and male sex were associated with severe outcomes (hospitalization or death). Persons at greater risk for severe outcomes should continue to take precautions to prevent SARS-CoV-2 infection, even if fully vaccinated.
Collapse
Affiliation(s)
- Brandy Darby
- Virginia Department of Health, 109 Governor St., Richmond, VA 23219, USA.
| | - Victoria Alexander
- Virginia Department of Health, 109 Governor St., Richmond, VA 23219, USA.
| | - Julia Murphy
- Virginia Department of Health, 109 Governor St., Richmond, VA 23219, USA.
| |
Collapse
|
539
|
Algaba A, Romero S, Granja A, Garza D, Aller M, Barrero S, Guerra I, Gil M, Pizarro N, Ruiz P, Prieto S, Hernández B, Pou A, Bermejo F. Serological response to vaccines against SARS-CoV-2 in patients with inflammatory bowel disease. GASTROENTEROLOGÍA Y HEPATOLOGÍA (ENGLISH EDITION) 2023. [PMCID: PMC9910024 DOI: 10.1016/j.gastre.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Objective To study the serological response (SR) and tolerability of COVID-19 vaccine in patients with inflammatory bowel disease (IBD) and its relation with IBD treatment and type of vaccine. Methods Observational, cross-sectional study in patients with IBD vaccinated against COVID-19 without known previous infection. SR was analyzed by the determination of IgG antibodies against the S1 subunit. Safety was studied using a questionnaire to identify adverse effects (AE). Results 280 patients with IBD were included. Type of vaccines: Comirnaty® 68.8%; Spikevax® 10.8%, Vaxzevria® 18.3%, Ad26.COV2-S® 2.2%. 51.3% had AE, being 100% mild. 65% developed IgG antibodies after vaccination. The SR was higher for vaccines with mRNA technology (100% Spikevax®, 68.5% Comirnaty®) compared to those based on adenovirus vector (38.0% Vaxzevria®, 33.3% Ad26.COV2-S®) (P < .001). In the multivariate analysis, SR was related to age (<60 years; OR: 3.8, 95% CI 1.9–7.0; P < .001). The SR in patients with aminosalicylates was 65.4%, 61.4% with immunosuppressants, 65.8% with anti-TNF, and 68.7% with non-anti-TNF biologicals (P = .9). Conclusions One third of patients with IBD did not develop antibodies with the initial vaccination against SARS-CoV-2. The SR to vaccines based on mRNA technology was higher, and it was related to age (higher in younger patients). Immunosuppressants and biologicals did not decrease SR. More than half of the patients presented AD, being mild in all cases.
Collapse
Affiliation(s)
- Alicia Algaba
- Servicio de Digestivo, Hospital Universitario de Fuenlabrada, Madrid, Spain,Corresponding author
| | - Sara Romero
- Servicio de Digestivo, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Alicia Granja
- Servicio de Digestivo, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Daniel Garza
- Servicio de Digestivo, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Mar Aller
- Servicio de Digestivo, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Sara Barrero
- Servicio de Digestivo, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Iván Guerra
- Servicio de Digestivo, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Marina Gil
- Servicio de Digestivo, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Nazaret Pizarro
- Servicio de Digestivo, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Paloma Ruiz
- Servicio de Digestivo, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Santiago Prieto
- Servicio de Laboratorio, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Belén Hernández
- Servicio de Farmacia, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Aranzazu Pou
- Servicio de Farmacia, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Fernando Bermejo
- Servicio de Digestivo, Hospital Universitario de Fuenlabrada, Madrid, Spain
| |
Collapse
|
540
|
Heath PT, Galiza EP, Baxter DN, Boffito M, Browne D, Burns F, Chadwick DR, Clark R, Cosgrove CA, Galloway J, Goodman AL, Heer A, Higham A, Iyengar S, Jeanes C, Kalra PA, Kyriakidou C, Bradley JM, Munthali C, Minassian AM, McGill F, Moore P, Munsoor I, Nicholls H, Osanlou O, Packham J, Pretswell CH, San Francisco Ramos A, Saralaya D, Sheridan RP, Smith R, Soiza RL, Swift PA, Thomson EC, Turner J, Viljoen ME, Fries L, Cho I, McKnight I, Glenn G, Rivers EJ, Robertson A, Alves K, Smith K, Toback S. Safety and Efficacy of the NVX-CoV2373 Coronavirus Disease 2019 Vaccine at Completion of the Placebo-Controlled Phase of a Randomized Controlled Trial. Clin Infect Dis 2023; 76:398-407. [PMID: 36210481 PMCID: PMC9619635 DOI: 10.1093/cid/ciac803] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/19/2022] [Accepted: 09/29/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The recombinant protein-based vaccine, NVX-CoV2373, demonstrated 89.7% efficacy against coronavirus disease 2019 (COVID-19) in a phase 3, randomized, observer-blinded, placebo-controlled trial in the United Kingdom. The protocol was amended to include a blinded crossover. Data to the end of the placebo-controlled phase are reported. METHODS Adults aged 18-84 years received 2 doses of NVX-CoV2373 or placebo (1:1) and were monitored for virologically confirmed mild, moderate, or severe COVID-19 (onset from 7 days after second vaccination). Participants who developed immunoglobulin G (IgG) against nucleocapsid protein but did not show symptomatic COVID-19 were considered asymptomatic. Secondary outcomes included anti-spike (S) IgG responses, wild-type virus neutralization, and T-cell responses. RESULTS Of 15 185 participants, 13 989 remained in the per-protocol efficacy population (6989 NVX-CoV2373, 7000 placebo). At a maximum of 7.5 months (median, 4.5) postvaccination, there were 24 cases of COVID-19 among NVX-CoV2373 recipients and 134 cases among placebo recipients, a vaccine efficacy of 82.7% (95% confidence interval [CI], 73.3%-88.8%). Vaccine efficacy was 100% (95% CI, 17.9%-100.0%) against severe disease and 76.3% (95% CI, 57.4%-86.8%) against asymptomatic disease. High anti-S and neutralization responses to vaccination were evident, together with S-protein-specific induction of interferon-γ secretion in peripheral blood T cells. Incidence of serious adverse events and adverse events of special interest were similar between groups. CONCLUSIONS A 2-dose regimen of NVX-CoV2373 conferred a high level of ongoing protection against asymptomatic, symptomatic, and severe COVID-19 through >6 months postvaccination. A gradual decrease of protection suggests that a booster may be indicated. CLINICAL TRIALS REGISTRATION EudraCT, 2020-004123-16.
Collapse
Affiliation(s)
- Paul T Heath
- Vaccine Institute and Paediatric Infectious Disease Research Group, St. George's, University of London and St. George's University Hospitals National Health Service Foundation Trust, London, United Kingdom
| | - Eva P Galiza
- Vaccine Institute, St. George's, University of London and St. George's University Hospitals National Health Service Foundation Trust, London, United Kingdom
| | - David Neil Baxter
- Medical Education, Stockport National Health Service Foundation Trust, Stepping Hill Hospital, Poplar Grove, Stockport, United Kingdom
| | - Marta Boffito
- Chelsea and Westminster Hospital National Health Service Foundation Trust and Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Duncan Browne
- Endocrinology/Diabetes/General Medicine, Royal Cornwall Hospitals National Health Service Trust, Truro, United Kingdom
| | - Fiona Burns
- Faculty of Population Health Sciences, Institute for Global Health, University College London, and Royal Free London National Health Service Foundation Trust, London, United Kingdom
| | - David R Chadwick
- Centre for Clinical Infection, South Tees Hospitals National Health Service Foundation Trust, James Cook University Hospital, Middlesbrough, United Kingdom
| | | | - Catherine A Cosgrove
- Vaccine Institute, St. George's, University of London and St. George's University Hospitals National Health Service Foundation Trust, London, United Kingdom
| | - James Galloway
- Centre for Rheumatic Disease, Kings College London, London, United Kingdom
| | - Anna L Goodman
- Department of Infectious Diseases, Guy's and St Thomas' National Health Service Foundation Trust, and Medical Research Council Clinical Trials Unit at University College London, London, United Kingdom
| | - Amardeep Heer
- Lakeside Healthcare Research, Lakeside Surgeries Corby, Northants, United Kingdom
| | - Andrew Higham
- Gastrointestinal and Liver Services, University Hospitals of Morecambe Bay National Health Service Foundation Trust, Kendal, United Kingdom
| | - Shalini Iyengar
- Accelerated Enrollment Solutions, Synexus Hexham Dedicated Research Site, Hexham General Hospital, Hexham, United Kingdom
| | - Christopher Jeanes
- Department of Microbiology, Norfolk and Norwich University Hospitals National Health Service Foundation Trust, Norwich, Norfolk, United Kingdom
| | - Philip A Kalra
- Nephrology, Salford Royal Hospital, Northern Care Alliance National Health Service Foundation Trust, Salford, United Kingdom
| | - Christina Kyriakidou
- Accelerated Enrollment Solutions, Synexus Midlands Dedicated Research Site, Birmingham Research Park, Birmingham, United Kingdom
| | - Judy M Bradley
- Dentistry and Biomedical Sciences, School of Medicine, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast, Northern Ireland, United Kingdom
| | - Chigomezgo Munthali
- Accelerated Enrollment Solutions, Synexus Merseyside Dedicated Research Site, Burlington House, Waterloo, Liverpool, United Kingdom
| | - Angela M Minassian
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, and Oxford Health National Health Service Foundation Trust, Warneford Hospital, Oxford, United Kingdom
| | - Fiona McGill
- Microbiology, Leeds Teaching Hospitals National Health Service Trust, Leeds, United Kingdom
| | - Patrick Moore
- The Adam Practice, Poole, Dorset, United Kingdom
- University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | - Imrozia Munsoor
- Accelerated Enrollment Solutions, Synexus Glasgow Dedicated Research Site, Venture Building, Kelvin Campus, Glasgow, Scotland, United Kingdom
| | - Helen Nicholls
- Accelerated Enrollment Solutions, Synexus Wales Dedicated Research Site, Riverside Court Gwaelod-y-Garth, Cardiff, Wales, United Kingdom
| | - Orod Osanlou
- School of Medical Sciences (Pharmacology/Pharmacy), Bangor University, and Clinical Pharmacology and Therapeutics/General Internal Medicine, Betsi Cadwaladr University Health Board, Wales, United Kingdom
| | - Jonathan Packham
- Academic Unit of Population and Lifespan Sciences, University of Nottingham, Nottingham, United Kingdom
- Rheumatology Department, Haywood Hospital, Midlands Partnership National Health Service Foundation Trust, Stafford, United Kingdom
| | - Carol H Pretswell
- Accelerated Enrollment Solutions, Synexus Lancashire Dedicated Research Site, Matrix Park Buckshaw Village, Chorley, Lancashire, United Kingdom
| | - Alberto San Francisco Ramos
- Vaccine Institute, St. George's, University of London and St. George's University Hospitals National Health Service Foundation Trust, London, United Kingdom
| | - Dinesh Saralaya
- National Institute for Health Research Patient Recruitment Centre and Bradford Teaching Hospitals National Health Service Foundation Trust, Bradford, United Kingdom
| | - Ray P Sheridan
- Geriatric Medicine, Royal Devon & Exeter Hospital, Exeter, Devon, United Kingdom
| | - Richard Smith
- Nephrology, East Suffolk and North Essex National Health Service Foundation Trust and University of Essex, Wivenhoe Park, Colchester, Essex, United Kingdom
| | - Roy L Soiza
- Aberdeen Royal Infirmary and Ageing Clinical and Experimental Research Group, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Pauline A Swift
- Renal Services, Epsom and St Helier University Hospitals National Health Service Trust, London, United Kingdom
| | - Emma C Thomson
- School of Infection & Immunity, Medical Research Council-University of Glasgow Centre for Virus Research, and Queen Elizabeth University Hospital, National Health Service Greater Glasgow & Clyde, Glasgow, Scotland, United Kingdom
| | - Jeremy Turner
- Diabetes and Endocrinology, Norfolk and Norwich University Hospitals National Health Service Foundation Trust, Norwich, Norfolk, United Kingdom
| | - Marianne Elizabeth Viljoen
- Accelerated Enrollment Solutions, Synexus Manchester Dedicated Research Site, Kilburn House, Manchester, United Kingdom
| | - Louis Fries
- Clinical Immunology, Novavax, Inc, Gaithersburg, Maryland, USA
| | - Iksung Cho
- Biostatistics, Novavax, Inc, Gaithersburg, Maryland, USA
| | - Irene McKnight
- Medical Writing, Novavax, Inc, Gaithersburg, Maryland, USA
| | - Greg Glenn
- Discovery, Novavax, Inc, Gaithersburg, Maryland, USA
| | - E Joy Rivers
- Clinical Development, Novavax, Inc, Gaithersburg, Maryland, USA
| | | | - Katia Alves
- Clinical Development, Novavax, Inc, Gaithersburg, Maryland, USA
| | - Kathy Smith
- Global Vaccine Safety, Novavax, Inc, Gaithersburg, Maryland, USA
| | - Seth Toback
- Medical Affairs, Novavax, Inc, Gaithersburg, Maryland, USA
| |
Collapse
|
541
|
Klaassen F, Chitwood MH, Cohen T, Pitzer VE, Russi M, Swartwood NA, Salomon JA, Menzies NA. Population Immunity to Pre-Omicron and Omicron Severe Acute Respiratory Syndrome Coronavirus 2 Variants in US States and Counties Through 1 December 2021. Clin Infect Dis 2023; 76:e350-e359. [PMID: 35717642 PMCID: PMC9214178 DOI: 10.1093/cid/ciac438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/20/2022] [Accepted: 05/28/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Both severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and coronavirus disease 2019 (COVID-19) vaccination contribute to population-level immunity against SARS-CoV-2. This study estimated the immunological exposure and effective protection against future SARS-CoV-2 infection in each US state and county over 2020-2021 and how this changed with the introduction of the Omicron variant. METHODS We used a Bayesian model to synthesize estimates of daily SARS-CoV-2 infections, vaccination data and estimates of the relative rates of vaccination conditional on infection status to estimate the fraction of the population with (1) immunological exposure to SARS-CoV-2 (ever infected with SARS-CoV-2 and/or received ≥1 doses of a COVID-19 vaccine), (2) effective protection against infection, and (3) effective protection against severe disease, for each US state and county from 1 January 2020 to 1 December 2021. RESULTS The estimated percentage of the US population with a history of SARS-CoV-2 infection or vaccination as of 1 December 2021 was 88.2% (95% credible interval [CrI], 83.6%-93.5%). Accounting for waning and immune escape, effective protection against the Omicron variant on 1 December 2021 was 21.8% (95% CrI, 20.7%-23.4%) nationally and ranged between 14.4% (13.2%-15.8%; West Virginia) and 26.4% (25.3%-27.8%; Colorado). Effective protection against severe disease from Omicron was 61.2% (95% CrI, 59.1%-64.0%) nationally and ranged between 53.0% (47.3%-60.0%; Vermont) and 65.8% (64.9%-66.7%; Colorado). CONCLUSIONS While more than four-fifths of the US population had prior immunological exposure to SARS-CoV-2 via vaccination or infection on 1 December 2021, only a fifth of the population was estimated to have effective protection against infection with the immune-evading Omicron variant.
Collapse
Affiliation(s)
- Fayette Klaassen
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Melanie H Chitwood
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Ted Cohen
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Virginia E Pitzer
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Marcus Russi
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Nicole A Swartwood
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Joshua A Salomon
- Department of Health Policy, Stanford University School of Medicine, Stanford, California, USA
| | - Nicolas A Menzies
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
542
|
Chinta S, Rodriguez-Guerra M, Shaban M, Pandey N, Jaquez-Duran M, Vittorio TJ. COVID-19 therapy and vaccination: a clinical narrative review. Drugs Context 2023; 12:2022-7-2. [PMID: 36793450 PMCID: PMC9914077 DOI: 10.7573/dic.2022-7-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 01/05/2023] [Indexed: 02/08/2023] Open
Abstract
The SARS-CoV-2 pandemic is the most globally impacting health issue our world has faced over the last century. As of January 7, 2022, around 300 million cases have been reported worldwide, with over 5 million deaths. The SARS-CoV-2 infection causes a hyperactive host immune response leading to an excessive inflammatory reaction with the release of many cytokines - cytokine storm - commonly noticed in acute respiratory distress syndrome, sepsis and fulminant multiorgan failure. Since the beginning of the pandemic, the scientific medical community has worked on therapeutic procedures that interfere with the exaggerated immune response. Thromboembolic complications are widespread in patients who are critically ill with COVID-19. Anticoagulant therapy was initially considered a cornerstone in hospitalized patients and even in the early post-discharge period; however, later trials have aborted the clinical benefits except for suspicion of or confirmed thrombosis. Immunomodulatory therapies are still crucial in moderate to severe COVID-19. Immunomodulator therapies include various medications from steroids to hydroxychloroquine, tocilizumab and Anakinra. Anti-inflammatory agents, vitamin supplements and antimicrobial therapy had initial encouraging evidence, but there are limited data to review. Convalescent plasma, immunoglobulins, eculizumab, neutralizing IgG1 monoclonal antibodies and remdesivir have positively impacted inpatient mortality and hospital length of stay. Eventually, wide population vaccination was proven to be the best tool to overcome the SARS-CoV-2 pandemic and help humanity return to regular life. Many vaccines and various strategies have been used since December 2020. This review discusses how the SARS-CoV-2 pandemic has progressed and surged, and summarizes the safety and efficacy of the most used therapies and vaccines in the light of recent evidence.
Collapse
Affiliation(s)
- Siddharth Chinta
- Department of Medicine, BronxCare Hospital Center, Icahn School of Medicine at Mt. Sinai, Bronx, NY, USA
| | - Miguel Rodriguez-Guerra
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mohammed Shaban
- Department of Medicine, BronxCare Hospital Center, Icahn School of Medicine at Mt. Sinai, Bronx, NY, USA
| | - Neelanjana Pandey
- Department of Medicine, BronxCare Hospital Center, Icahn School of Medicine at Mt. Sinai, Bronx, NY, USA
| | - Maria Jaquez-Duran
- Department of Medicine, BronxCare Hospital Center, Icahn School of Medicine at Mt. Sinai, Bronx, NY, USA
| | - Timothy J Vittorio
- Division of Cardiology, BronxCare Hospital Center, Icahn School of Medicine at Mt. Sinai, Bronx, NY, USA
| |
Collapse
|
543
|
Fritsche LG, Jin W, Admon AJ, Mukherjee B. Characterizing and Predicting Post-Acute Sequelae of SARS CoV-2 Infection (PASC) in a Large Academic Medical Center in the US. J Clin Med 2023; 12:1328. [PMID: 36835863 PMCID: PMC9967320 DOI: 10.3390/jcm12041328] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/30/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND A growing number of Coronavirus Disease-2019 (COVID-19) survivors are affected by post-acute sequelae of SARS CoV-2 infection (PACS). Using electronic health record data, we aimed to characterize PASC-associated diagnoses and develop risk prediction models. METHODS In our cohort of 63,675 patients with a history of COVID-19, 1724 (2.7%) had a recorded PASC diagnosis. We used a case-control study design and phenome-wide scans to characterize PASC-associated phenotypes of the pre-, acute-, and post-COVID-19 periods. We also integrated PASC-associated phenotypes into phenotype risk scores (PheRSs) and evaluated their predictive performance. RESULTS In the post-COVID-19 period, known PASC symptoms (e.g., shortness of breath, malaise/fatigue) and musculoskeletal, infectious, and digestive disorders were enriched among PASC cases. We found seven phenotypes in the pre-COVID-19 period (e.g., irritable bowel syndrome, concussion, nausea/vomiting) and sixty-nine phenotypes in the acute-COVID-19 period (predominantly respiratory, circulatory, neurological) associated with PASC. The derived pre- and acute-COVID-19 PheRSs stratified risk well, e.g., the combined PheRSs identified a quarter of the cohort with a history of COVID-19 with a 3.5-fold increased risk (95% CI: 2.19, 5.55) for PASC compared to the bottom 50%. CONCLUSIONS The uncovered PASC-associated diagnoses across categories highlighted a complex arrangement of presenting and likely predisposing features, some with potential for risk stratification approaches.
Collapse
Affiliation(s)
- Lars G. Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Precision Health Data Science, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Weijia Jin
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Precision Health Data Science, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Andrew J. Admon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- VA Center for Clinical Management Research, LTC Charles S. Kettles VA Medical Center, Ann Arbor, MI 48109, USA
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Precision Health Data Science, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
544
|
Ozhmegova EN, Savochkina TE, Prilipov AG, Tikhomirov E, Larichev VF, Sayfullin MA, Grebennikova TV. [Molecular epidemiological analysis of SARS-CoV-2 genovariants in Moscow and Moscow region]. Vopr Virusol 2023; 67:496-505. [PMID: 37264839 DOI: 10.36233/0507-4088-146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Indexed: 06/03/2023]
Abstract
INTRODUCTION SARS-CoV-2, a severe acute respiratory illness virus that emerged in China in late 2019, continues to spread rapidly around the world, accumulating mutations and thus causing serious concern. Five virus variants of concern are currently known: Alpha (lineage B.1.1.7), Beta (lineage B.1.351), Gamma (lineage P.1), Delta (lineage B.1.617.2), and Omicron (lineage B.1.1.529). In this study, we conducted a molecular epidemiological analysis of the most prevalent genovariants in Moscow and the region. The aim of the study is to estimate the distribution of various variants of SARS-CoV-2 in Moscow city and the Moscow Region. MATERIALS AND METHODS 227 SARS-CoV-2 sequences were used for analysis. Isolation of the SARS-CoV-2 virus was performed on Vero E6 cell culture. Sequencing was performed by the Sanger method. Bioinformatic analysis was carried out using software packages: MAFFT, IQ-TREE v1.6.12, jModelTest 2.1.7, Nextstrain, Auspice v2.34. RESULTS As a result of phylogenetic analysis, we have identified the main variants of the virus circulating in Russia that have been of concern throughout the existence of the pandemic, namely: variant B.1.1.7, which accounted for 30% (9/30), AY.122, which accounted for 16.7% (5/30), BA.1.1 with 20% (6/30) and B.1.1 with 33.3% (10/30). When examining Moscow samples for the presence of mutations in SARS-CoV-2 structural proteins of different genovariants, a significant percentage of the most common substitutions was recorded: S protein D614G (86.7%), P681H/R (63.3%), E protein T9I (20.0%); M protein I82T (30.0%), D3G (20.0%), Q19E (20.0%) and finally N protein R203K/M (90.0%), G204R/P (73.3 %). CONCLUSION The study of the frequency and impact of mutations, as well as the analysis of the predominant variants of the virus are important for the development and improvement of vaccines for the prevention of COVID-19. Therefore, ongoing molecular epidemiological studies are needed, as these data provide important information about changes in the genome of circulating SARS-CoV-2 variants.
Collapse
Affiliation(s)
- E N Ozhmegova
- National Research Center for Epidemiology and Microbiology named after honorary academician N.F. Gamaleya, Ministry of Health of the Russian Federation
| | - T E Savochkina
- National Research Center for Epidemiology and Microbiology named after honorary academician N.F. Gamaleya, Ministry of Health of the Russian Federation
| | - A G Prilipov
- National Research Center for Epidemiology and Microbiology named after honorary academician N.F. Gamaleya, Ministry of Health of the Russian Federation
| | - E Tikhomirov
- National Research Center for Epidemiology and Microbiology named after honorary academician N.F. Gamaleya, Ministry of Health of the Russian Federation
| | - V F Larichev
- National Research Center for Epidemiology and Microbiology named after honorary academician N.F. Gamaleya, Ministry of Health of the Russian Federation
| | - M A Sayfullin
- National Research Center for Epidemiology and Microbiology named after honorary academician N.F. Gamaleya, Ministry of Health of the Russian Federation
- Pirogov Russian National Research Medical University
| | - T V Grebennikova
- National Research Center for Epidemiology and Microbiology named after honorary academician N.F. Gamaleya, Ministry of Health of the Russian Federation
| |
Collapse
|
545
|
Qiao Y, Wotring JW, Zhang CJ, Jiang X, Xiao L, Watt A, Gattis D, Scandalis E, Freier S, Zheng Y, Pretto CD, Ellison SJ, Swayze EE, Guo S, Sexton JZ, Chinnaiyan AM. Antisense oligonucleotides to therapeutically target SARS-CoV-2 infection. PLoS One 2023; 18:e0281281. [PMID: 36735698 PMCID: PMC9897518 DOI: 10.1371/journal.pone.0281281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/19/2023] [Indexed: 02/04/2023] Open
Abstract
Although the COVID-19 pandemic began over three years ago, the virus responsible for the disease, SARS-CoV-2, continues to infect people across the globe. As such, there remains a critical need for development of novel therapeutics against SARS-CoV-2. One technology that has remained relatively unexplored in COVID-19 is the use of antisense oligonucleotides (ASOs)-short single-stranded nucleic acids that bind to target RNA transcripts to modulate their expression. In this study, ASOs targeted against the SARS-CoV-2 genome and host entry factors, ACE2 and TMPRSS2, were designed and tested for their ability to inhibit cellular infection by SARS-CoV-2. Using our previously developed SARS-CoV-2 bioassay platform, we screened 180 total ASOs targeting various regions of the SARS-CoV-2 genome and validated several ASOs that potently blocked SARS-CoV-2 infection in vitro. Notably, select ASOs retained activity against both the WA1 and B.1.1.7 (commonly known as alpha) variants. Screening of ACE2 and TMPRSS2 ASOs showed that targeting of ACE2 also potently prevented infection by the WA1 and B.1.1.7 SARS-CoV-2 viruses in the tested cell lines. Combined with the demonstrated success of ASOs in other disease indications, these results support further research into the development of ASOs targeting SARS-CoV-2 and host entry factors as potential COVID-19 therapeutics.
Collapse
Affiliation(s)
- Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, United States of America
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States of America
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Jesse W. Wotring
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States of America
| | - Charles J. Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States of America
| | - Xia Jiang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, United States of America
| | - Lanbo Xiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, United States of America
| | - Andy Watt
- Ionis Pharmaceuticals, Carlsbad, CA, United States of America
| | - Danielle Gattis
- Ionis Pharmaceuticals, Carlsbad, CA, United States of America
| | - Eli Scandalis
- Ionis Pharmaceuticals, Carlsbad, CA, United States of America
| | - Susan Freier
- Ionis Pharmaceuticals, Carlsbad, CA, United States of America
| | - Yang Zheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, United States of America
| | - Carla D. Pretto
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Stephanie J. Ellison
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, United States of America
| | - Eric E. Swayze
- Ionis Pharmaceuticals, Carlsbad, CA, United States of America
| | - Shuling Guo
- Ionis Pharmaceuticals, Carlsbad, CA, United States of America
| | - Jonathan Z. Sexton
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States of America
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
- Center for Drug Repurposing, University of Michigan, Ann Arbor, MI, United States of America
- Michigan Institute for Clinical and Health Research, University of Michigan, Ann Arbor, MI, United States of America
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, United States of America
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States of America
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States of America
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, United States of America
- Department of Urology, University of Michigan, Ann Arbor, MI, United States of America
| |
Collapse
|
546
|
Martínez-Baz I, Trobajo-Sanmartín C, Miqueleiz A, Casado I, Navascués A, Burgui C, Ezpeleta C, Castilla J, Guevara M, the Working Group for the Study of COVID-19 in Navarra. Risk reduction of hospitalisation and severe disease in vaccinated COVID-19 cases during the SARS-CoV-2 variant Omicron BA.1-predominant period, Navarre, Spain, January to March 2022. Euro Surveill 2023; 28:2200337. [PMID: 36729113 PMCID: PMC9896606 DOI: 10.2807/1560-7917.es.2023.28.5.2200337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BackgroundAs COVID-19 vaccine effectiveness against SARS-CoV-2 infection was lower for cases of the Omicron vs the Delta variant, understanding the effect of vaccination in reducing risk of hospitalisation and severe disease among COVID-19 cases is crucial.AimTo evaluate risk reduction of hospitalisation and severe disease in vaccinated COVID-19 cases during the Omicron BA.1-predominant period in Navarre, Spain.MethodsA case-to-case comparison included COVID-19 epidemiological surveillance data in adults ≥ 18 years from 3 January-20 March 2022. COVID-19 vaccination status was compared between hospitalised and non-hospitalised cases, and between severe (intensive care unit admission or death) and non-severe cases using logistic regression models.ResultsAmong 58,952 COVID-19 cases, 565 (1.0%) were hospitalised and 156 (0.3%) were severe. The risk of hospitalisation was reduced within the first 6 months after full COVID-19 vaccination (complete primary series) (adjusted odds ratio (aOR): 0.06; 95% CI: 0.04-0.09) and after 6 months (aOR: 0.16; 95% CI: 0.12-0.21; pcomparison < 0.001), as well as after a booster dose (aOR: 0.06: 95% CI: 0.04-0.07). Similarly, the risk of severe disease was reduced (aOR: 0.13, 0.18, and 0.06, respectively). Compared with cases fully vaccinated 6 months or more before a positive test, those who had received a booster dose had lower risk of hospitalisation (aOR: 0.38; 95% CI: 0.28-0.52) and severe disease (aOR: 0.38; 95% CI: 0.21-0.68).ConclusionsFull COVID-19 vaccination greatly reduced the risk of hospitalisation and severe outcomes in COVID-19 cases with the Omicron variant, and a booster dose improved this effect in people aged over 65 years.
Collapse
Affiliation(s)
- Iván Martínez-Baz
- Instituto de Salud Pública de Navarra, Pamplona, Spain,CIBER Epidemiología y Salud Pública (CIBERESP), Pamplona, Spain,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Camino Trobajo-Sanmartín
- Instituto de Salud Pública de Navarra, Pamplona, Spain,CIBER Epidemiología y Salud Pública (CIBERESP), Pamplona, Spain,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Ana Miqueleiz
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain,Clinical Microbiology Department, Hospital Universitario de Navarra, Pamplona, Spain
| | - Itziar Casado
- Instituto de Salud Pública de Navarra, Pamplona, Spain,CIBER Epidemiología y Salud Pública (CIBERESP), Pamplona, Spain,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Ana Navascués
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain,Clinical Microbiology Department, Hospital Universitario de Navarra, Pamplona, Spain
| | - Cristina Burgui
- Instituto de Salud Pública de Navarra, Pamplona, Spain,CIBER Epidemiología y Salud Pública (CIBERESP), Pamplona, Spain,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Carmen Ezpeleta
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain,Clinical Microbiology Department, Hospital Universitario de Navarra, Pamplona, Spain
| | - Jesús Castilla
- Instituto de Salud Pública de Navarra, Pamplona, Spain,CIBER Epidemiología y Salud Pública (CIBERESP), Pamplona, Spain,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Marcela Guevara
- Instituto de Salud Pública de Navarra, Pamplona, Spain,CIBER Epidemiología y Salud Pública (CIBERESP), Pamplona, Spain,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | | | | |
Collapse
|
547
|
Zacharopoulou E, Orfanoudaki E, Tzouvala M, Tribonias G, Kokkotis G, Kitsou V, Almpani F, Christidou A, Viazis N, Mantzaris GJ, Tsafaridou M, Karmiris K, Theodoropoulou A, Papathanasiou E, Zampeli E, Michopoulos S, Tigkas S, Michalopoulos G, Laoudi E, Karatzas P, Mylonas I, Kyriakos N, Liatsos C, Kafetzi T, Theocharis G, Taka S, Panagiotopoulou K, Koutroubakis IE, Bamias G. Patients With Inflammatory Bowel Diseases Have Impaired Antibody Production After Anti-SARS-CoV-2 Vaccination: Results From a Panhellenic Registry. Inflamm Bowel Dis 2023; 29:228-237. [PMID: 35394529 PMCID: PMC9051580 DOI: 10.1093/ibd/izac068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Four EMA-approved vaccines against SARS-CoV-2 are currently available. Data regarding antibody responses to initial vaccination regimens in patients with inflammatory bowel diseases (IBD) are limited. METHODS We conducted a prospective, controlled, multicenter study in tertiary Greek IBD centers. Participating patients had completed the initial vaccination regimens (1 or 2 doses, depending on the type of COVID-19 vaccine) at least 2 weeks before study enrolment. Anti-S1 IgG antibody levels were measured. Demographic and adverse events data were collected. RESULTS We tested 403 patients (Crohn's disease, 58.9%; male, 53.4%; median age, 45 years) and 124 healthy controls (HCs). Following full vaccination, 98% of patients seroconverted, with mRNA vaccines inducing higher seroconversion rates than viral vector vaccines (P = .021). In total, IBD patients had lower anti-S1 levels than HCs (P < .001). In the multivariate analysis, viral vector vaccines (P < .001), longer time to antibody testing (P < .001), anti-TNFα treatment (P = .013), and age (P = .016) were independently associated with lower anti-S1 titers. Vedolizumab monotherapy was associated with higher antibody levels than anti-TNFα or anti-interleukin-12/IL-23 monotherapy (P = .023 and P = .032). All anti- SARS-CoV-2 vaccines were safe. CONCLUSIONS Patients with IBD have impaired antibody responses to anti-SARS-CoV-2 vaccination, particularly those receiving viral vector vaccines and those on anti-TNFα treatment. Older age also hampers antibody production after vaccination. For those low-response groups, administration of accelerated or prioritized booster vaccination may be considered.
Collapse
Affiliation(s)
- Eirini Zacharopoulou
- Department of Gastroenterology, General Hospital of Nikaia Piraeus “Agios Panteleimon,” General Hospital Dytikis Attikis “Agia Varvara,”Athens, Greece
| | - Eleni Orfanoudaki
- Department of Gastroenterology, University Hospital of Heraklion, Medical School, University of Crete, Heraklion Crete, Greece
| | - Maria Tzouvala
- Department of Gastroenterology, General Hospital of Nikaia Piraeus “Agios Panteleimon,” General Hospital Dytikis Attikis “Agia Varvara,”Athens, Greece
| | - George Tribonias
- Department of Gastroenterology, General Hospital of Nikaia Piraeus “Agios Panteleimon,” General Hospital Dytikis Attikis “Agia Varvara,”Athens, Greece
| | - Georgios Kokkotis
- GI Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian Univeristy of Athens- “Sotiria” General Hospital, Athens, Greece
| | - Vassiliki Kitsou
- GI Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian Univeristy of Athens- “Sotiria” General Hospital, Athens, Greece
| | - Foteini Almpani
- Department of Gastroenterology, General Hospital of Athens, Evaggelismos, Opthalmiatreion Athinon, Polykliniki, Athens, Greece
| | - Aggeliki Christidou
- Department of Gastroenterology, General Hospital of Athens, Evaggelismos, Opthalmiatreion Athinon, Polykliniki, Athens, Greece
| | - Nikolaos Viazis
- Department of Gastroenterology, General Hospital of Athens, Evaggelismos, Opthalmiatreion Athinon, Polykliniki, Athens, Greece
| | - Gerassimos J Mantzaris
- Department of Gastroenterology, General Hospital of Athens, Evaggelismos, Opthalmiatreion Athinon, Polykliniki, Athens, Greece
| | - Maria Tsafaridou
- Department of Gastroenterology, University Hospital of Heraklion, Medical School, University of Crete, Heraklion Crete, Greece
| | | | | | - Evgenia Papathanasiou
- Department of Gastroenterology, General Hospital of Athens “Alexandra,”Athens, Greece
| | - Evanthia Zampeli
- Department of Gastroenterology, General Hospital of Athens “Alexandra,”Athens, Greece
| | - Spyridon Michopoulos
- Department of Gastroenterology, General Hospital of Athens “Alexandra,”Athens, Greece
| | - Stefanos Tigkas
- **Department of Gastroenterology, General Hospital of Piraeus “Tzaneio,”Piraeus, Greece
| | | | - Efrossini Laoudi
- Department of Gastroenterology, National and Kapodistrian University of Athens, General Hospital of Athens “Laiko,”Athens, Greece
| | - Pantelis Karatzas
- Department of Gastroenterology, National and Kapodistrian University of Athens, General Hospital of Athens “Laiko,”Athens, Greece
| | - Iordanis Mylonas
- Department of Gastroenterology, 401 General Army Hospital of Athens, Athens, Greece
| | - Nikolaos Kyriakos
- Department of Gastroenterology, 401 General Army Hospital of Athens, Athens, Greece
| | - Christos Liatsos
- Department of Gastroenterology, 401 General Army Hospital of Athens, Athens, Greece
| | - Theodora Kafetzi
- Department of Gastroenterology, University Hospital of Patras, Patra, Greece
| | - Georgios Theocharis
- Department of Gastroenterology, University Hospital of Patras, Patra, Greece
| | - Styliani Taka
- StArtBio PC Molecular Diagnostics and Biotechnology Services, Athens, Greece
| | | | - Ioannis E Koutroubakis
- Department of Gastroenterology, University Hospital of Heraklion, Medical School, University of Crete, Heraklion Crete, Greece
| | - Giorgos Bamias
- GI Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian Univeristy of Athens- “Sotiria” General Hospital, Athens, Greece
| |
Collapse
|
548
|
Collie S, Saggers RT, Bandini R, Steenkamp L, Champion J, Gray G, Bekker LG, Goga A, Garrett N, Patricios J. Association between regular physical activity and the protective effect of vaccination against SARS-CoV-2 in a South African case-control study. Br J Sports Med 2023; 57:205-211. [PMID: 36280289 DOI: 10.1136/bjsports-2022-105734] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Both vaccination and physical activity have been shown to independently decrease the likelihood of severe COVID-19 infection. OBJECTIVE To assess the association between regular physical activity and vaccination against COVID-19 among healthcare workers. METHODS A test negative case-control study design was used to estimate the risk of having an associated COVID-19-related hospital admission, among individuals who were unvaccinated compared with those who were fully vaccinated with Ad26.COV2.S (>28 days after a single dose). 196 444 participant tests were stratified into three measured physical activity subgroups with low, moderate and high activity, to test the hypothesis that physical activity is an effect modifier on the relationship between vaccination and hospitalisation. RESULTS Vaccine effectiveness against a COVID-19-related admission among vaccinated individuals within the low activity group was 60.0% (95% CI 39.0 to 73.8), 72.1% (95% CI 55.2 to 82.6) for the moderate activity group, and 85.8% (95% CI 74.1 to 92.2) for the high activity group. Compared with individuals with low activity levels, vaccinated individuals with moderate and high activity levels had a 1.4 (95% CI 1.36 to 1.51) and 2.8 (95% CI 2.35 to 3.35) times lower risk of COVID-19 admission, respectively (p value <0.001 for both groups). CONCLUSIONS Regular physical activity was associated with improved vaccine effectiveness against COVID-19 hospitalisation, with higher levels of physical activity associated with greater vaccine effectiveness. Physical activity enhances vaccine effectiveness against severe COVID-19 outcomes and should be encouraged by greater public health messaging.
Collapse
Affiliation(s)
- Shirley Collie
- Healthcare Analytics, Discovery Health, Johannesburg, South Africa
| | - Robin Terence Saggers
- Wits Sport and Health (WiSH), School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa.,Department of Paediatrics and Child Health, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Rossella Bandini
- PRINCE: Project to Improve Neonatal Care, School of Clinical Medicine, Faculty of Health Sciences, Wits University, Johannesburg-Braamfontein, Gauteng, South Africa
| | | | - Jared Champion
- Healthcare Analytics, Discovery Health, Johannesburg, South Africa
| | - Glenda Gray
- COVID-19 Research Committee, South African Medical Research Council, Tygerberg, South Africa
| | - Linda-Gail Bekker
- Desmond Tutu HIV Centre, University of Cape Town, Observatory, South Africa
| | - Ameena Goga
- COVID-19 Research Committee, South African Medical Research Council, Tygerberg, South Africa.,Department of Paediatrics and Child Health, University of Pretoria, Pretoria, South Africa
| | - Nigel Garrett
- Centre for the Aids Programme of Research in South Africa, Durban, KwaZulu-Natal, South Africa.,School of Nursing and Public Health, Discipline of Public Health Medicine, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Jon Patricios
- Wits Sport and Health (WiSH), School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| |
Collapse
|
549
|
Han TY, Shim DH, Lee YJ, Lee YB, Koo HYR, Shin MK, Kim TE, Jang YH, Bang JS, Kook HD, Ahn J, Jung HJ, Na CH. Effect of COVID-19 (SARS-CoV-2) Vaccination on Patients with Atopic Dermatitis Treated with Dupilumab: A Multicenter, Observational Study. Ann Dermatol 2023; 35:38-45. [PMID: 36750457 PMCID: PMC9905853 DOI: 10.5021/ad.22.118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/26/2022] [Accepted: 10/12/2022] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Atopic dermatitis (AD) patients usually wonder if their condition will worsen after vaccination or if they should continue with the treatment they are receiving. Considering that many patients treated with dupilumab had previously experienced severe AD symptoms and flares, the concerns are more understandable. OBJECTIVE This study aimed to investigate the safety of the coronavirus disease 2019 (COVID-19) vaccination in patients with AD treated with dupilumab. METHODS We enrolled 133 patients (101 dupilumab-treated and 32 systemic oral agents-treated as control group) with AD from six hospitals. Patients were asked about worsening pruritus and AD (5-point Likert scale) after vaccination. AD variables (eczema area and severity index [EASI], investigator's global assessment [IGA], itch numerical rating scale [NRS], sleep NRS, and patient-oriented eczema measure [POEM]) were compared pre- and post-vaccination. Adverse reactions to the COVID-19 vaccination were observed. RESULTS The incidence of adverse reactions to COVID-19 vaccines and worsening AD symptoms in dupilumab-treated patients were not significantly different compared with that in the control group. The itch NRS score increased significantly after vaccination (p<0.001). However, there were no statistically significant differences between the pre-and post-EASI, IGA, and POEM scores. Eight patients (7.9%) had worse EASI scores and required rescue therapy; however, most were easily managed with low-dose steroids or topical agents. None of the patients discontinued dupilumab treatment. CONCLUSION No serious adverse reactions were observed in patients with AD after COVID-19 vaccination. Exacerbation of pruritus and AD symptoms was observed but was mostly mild and transient.
Collapse
Affiliation(s)
- Tae Young Han
- Department of Dermatology, College of Medicine, Eulji University, Daejeon, Korea
| | - Dong Hyun Shim
- Department of Dermatology, College of Medicine, Chosun University, Gwangju, Korea
| | - Yu Jin Lee
- Department of Dermatology, College of Medicine, Eulji University, Daejeon, Korea
| | - Young Bok Lee
- Department of Dermatology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ha Yeh Rin Koo
- Department of Dermatology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Min Kyung Shin
- Department of Dermatology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Tae Eun Kim
- Department of Dermatology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Yong Hyun Jang
- Department of Dermatology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jin Seon Bang
- Department of Dermatology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Hyung Don Kook
- Department of Dermatology, National Medical Center, Seoul, Korea
| | - Jiyoung Ahn
- Department of Dermatology, National Medical Center, Seoul, Korea
| | - Hye Jung Jung
- Department of Dermatology, National Medical Center, Seoul, Korea
| | - Chan Ho Na
- Department of Dermatology, College of Medicine, Chosun University, Gwangju, Korea.
| |
Collapse
|
550
|
Townsend JP, Hassler HB, Dornburg A. Infection by SARS-CoV-2 with alternate frequencies of mRNA vaccine boosting. J Med Virol 2023; 95:e28461. [PMID: 36602045 DOI: 10.1002/jmv.28461] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/16/2022] [Accepted: 12/26/2022] [Indexed: 01/06/2023]
Abstract
One of the most consequential unknowns of the COVID-19 pandemic is the frequency at which vaccine boosting provides sufficient protection from infection. We quantified the statistical likelihood of breakthrough infections over time following different boosting schedules with messenger RNA (mRNA)-1273 (Moderna) and BNT162b2 (Pfizer-BioNTech). We integrated anti-Spike IgG antibody optical densities with profiles of the waning of antibodies and corresponding probabilities of infection associated with coronavirus endemic transmission. Projecting antibody levels over time given boosting every 6 months, 1, 1.5, 2, or 3 years yielded respective probabilities of fending off infection over a 6-year span of >93%, 75%, 55%, 40%, and 24% (mRNA-1273) and >89%, 69%, 49%, 36%, and 23% (BNT162b2). Delaying the administration of updated boosters has bleak repercussions. It increases the probability of individual infection by SARS-CoV-2, and correspondingly, ongoing disease spread, prevalence, morbidity, hospitalization, and mortality. Instituting regular, population-wide booster vaccination updated to predominant variants has the potential to substantially forestall-and with global, widespread uptake, eliminate-COVID-19.
Collapse
Affiliation(s)
- Jeffrey P Townsend
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA.,Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut, USA.,Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, USA.,Program in Microbiology, Yale University, New Haven, Connecticut, USA
| | - Hayley B Hassler
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA
| | - Alex Dornburg
- Department of Bioinformatics and Genomics, University of North Carolina, Charlotte, North Carolina, USA
| |
Collapse
|