351
|
Rojas-Botero ML, Fernández-Niño JA, Arregocés-Castillo L, Palacios-Clavijo A, Pinto-Álvarez M, Ruiz-Gómez F. Real-world effectiveness of COVID-19 vaccines among Colombian adults: A retrospective, population-based study of the ESPERANZA cohort. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001845. [PMID: 37682804 PMCID: PMC10491003 DOI: 10.1371/journal.pgph.0001845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/14/2023] [Indexed: 09/10/2023]
Abstract
COVID-19 vaccines have been essential for reducing the impact of the pandemic; nevertheless, population-based data under real-life conditions are needed to compare their effectiveness in various contexts. The objective of this study was to estimate the effectiveness of vaccines in preventing hospitalization and death from COVID-19 in Colombia under real-life conditions among people aged 18 years and older, according to sex, age, confirmed history of COVID-19 and vaccination series, including the effects of boosters. This investigation was an observational, retrospective, population-based study based on the Colombian cohort "Esperanza". A total of 14,213,409 individuals aged 18 years and older were analyzed, who were matched in a 1:1 ratio of vaccinated to unvaccinated. The study groups consisted of unvaccinated individuals, those with a complete series (CS) and individuals with a CS plus booster. The vaccinated individuals received either homologous or heterologous vaccinations with Ad26.COV2-S, BNT162b2, ChAdOx1 nCoV-19, CoronaVac and mRNA-1273 vaccines. Follow-up was conducted between February 2021 and June 2022. Cox proportional hazards models were used, adjusted for potential confounders, to estimate the effectiveness of different vaccination series. For adults aged 18 years and older, the overall effectiveness of the vaccines in preventing hospitalization was 82.7% (95% CI 82.1-83.2) for CS and 80.2% (95%CI 78.7-81.6) for CS + booster. The effectiveness in preventing death was 86.0% (95%CI 85.5-86.5) for CS and 83.1% (95%CI 81.5-84.5) for CS + booster. Effectiveness decreased with age. While all efficacies were high, CoronaVac offered significantly lower protection, although this improved with a booster. Continued mass vaccination is pivotal, especially in low- and middle-income countries. The study highlights both the real-world effectiveness of these vaccines and the challenges in understanding waning immunity and the influence of different VoC(Variants of Concern) on results.
Collapse
Affiliation(s)
| | - Julián Alfredo Fernández-Niño
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Universidad del Norte, Barranquilla, Colombia
| | | | | | | | | |
Collapse
|
352
|
Angeletti A, Lugani F, La Porta E, Verrina E, Caridi G, Ghiggeri GM. Vaccines and nephrotic syndrome: efficacy and safety. Pediatr Nephrol 2023; 38:2915-2928. [PMID: 36512075 PMCID: PMC9745735 DOI: 10.1007/s00467-022-05835-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 12/15/2022]
Abstract
Vaccines represent the most important medical evolution in the last two centuries allowing prevention and formally eradication of a wide number of infectious diseases. Safety and effectiveness are main issues that still require an open discussion. A few clinical reports described a critical temporal relationship between vaccination and acute nephrotic syndrome, indirectly suggesting an association. For this review, the literature was reviewed to identify articles reporting associations of nephrotic syndrome with vaccines against a vast array of infectious diseases (including bacteria, virus and Sars-Cov-2). As specific aims, we evaluated effectiveness and safety in terms of occurrence of either "de novo" nephrotic syndrome in health subjects or "relapse" in those already affected by the disease. In total, 377 articles were found; 166 duplicates and 71 non-full text, animal studies or non-English language were removed. After excluding another 50 articles not containing relevant data on generic side effects or on relapses or new onset nephrotic syndrome, 90 articles met the search criteria. Overall, studies reported the effect of vaccines in 1015 patients, plus 4 nationwide epidemiologic investigations. Limited experience on vaccination of NS patients with measles, mumps, and rubella live attenuated vaccines does not allow any definitive conclusion on their safeness. VZV has been administered more frequently without side effects. Vaccines utilizing virus inactivated, recombinant, and toxoid can be utilized without risks in NS. Vaccines for influenza reduce the risk of infections during the pandemic and are associated with reduced risk of relapse of NS typically induced by the infection. Vaccines for SARS-CoV-2 (all kinds) offer a concrete approach to reduce the pandemic. "De novo" NS or recurrence are very rare and respond to common therapies.
Collapse
Affiliation(s)
- Andrea Angeletti
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy.
| | - Francesca Lugani
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Edoardo La Porta
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Enrico Verrina
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Gianluca Caridi
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Gian Marco Ghiggeri
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
353
|
Liang EC, Onstad LE, Carpenter P, Pergam SA, Flowers ME, Lee SJ, Liu C. Association of Self-Reported COVID-19 Vaccination Status with COVID-19 Infection among Adult Long-Term Hematopoietic Cell Transplantation Survivors. Transplant Cell Ther 2023; 29:584.e1-584.e9. [PMID: 37394113 PMCID: PMC10528463 DOI: 10.1016/j.jtct.2023.06.017] [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] [Received: 04/17/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/04/2023]
Abstract
Hematopoietic cell transplantation (HCT) recipients experience significant morbidity and mortality from coronavirus disease 19 (COVID-19) infection. Data are limited regarding long-term HCT survivors' uptake of and experiences with COVID-19 vaccination and infection. This study aimed to characterize COVID-19 vaccination uptake, use of other prevention measures, and COVID-19 infection outcomes in adult HCT recipients at our institution. Between July 1, 2021, and June 30, 2022, long-term adult HCT survivors were surveyed regarding overall health, chronic graft-versus-host (cGVHD) status, and experiences with COVID-19 vaccinations, prevention measures, and infections. Patients reported COVID-19 vaccination status, vaccine-related adverse effects, use of nonpharmaceutical prevention measures, and infections. Comparisons by response and vaccination status were performed using the chi-square test and Fisher exact test for categorical variables and the Kruskal-Wallis test for continuous variables. Of 4758 adult HCT survivors who underwent HCT between 1971 and 2021 and consented to participate in annual surveys, 1719 (36%) completed the COVID-19 module, and 1598 of 1705 (94%) reported receiving ≥1 dose of COVID-19 vaccine. Severe vaccine-related adverse effects were infrequent (5%). Among respondents receiving an mRNA vaccine, completion of doses according to the Centers for Disease Control and Prevention's vaccine recommendations at the time of survey return was 2 doses in 675 of 759 (89%), 3 doses in 610 of 778 (78%), and 4 doses in 26 of 55 (47%). Two hundred fifty respondents (15%) reported COVID-19 infection; 25 (10%) required hospitalization. Vaccinated respondents reported significantly higher uptake of household vaccination (1284 of 1404 [91%] versus 18 of 88 [20%]; P < .001) and the use of nonpharmaceutical interventions (P < .001). Vaccinated respondents were significantly less likely to have contracted COVID-19 (85 of 1480 [6%] versus 130 of 190 [68%]; P < .001), as were their household members (149 of 1451 [10%] versus 85 of 185 [46%]; P < .001). Receipt of additional COVID-19 vaccine doses beyond the first dose was associated with a reduced risk of COVID-19 infection (odds ratio, .63; 95% confidence interval, .47 to .85; P = .002). Vaccination was well tolerated and associated with a lower risk of COVID-19 infection among HCT survivors and their household contacts. Vaccination and booster doses should be encouraged as part of a multifaceted approach in this high-risk population.
Collapse
Affiliation(s)
- Emily C Liang
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Lynn E Onstad
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Paul Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Pediatrics, University of Washington, Seattle, Washington
| | - Steven A Pergam
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Mary E Flowers
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Stephanie J Lee
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Catherine Liu
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington.
| |
Collapse
|
354
|
Zhu X, Gebo KA, Abraham AG, Habtehyimer F, Patel EU, Laeyendecker O, Gniadek TJ, Fernandez RE, Baker OR, Ram M, Cachay ER, Currier JS, Fukuta Y, Gerber JM, Heath SL, Meisenberg B, Huaman MA, Levine AC, Shenoy A, Anjan S, Blair JE, Cruser D, Forthal DN, Hammitt LL, Kassaye S, Mosnaim GS, Patel B, Paxton JH, Raval JS, Sutcliffe CG, Abinante M, Broderick P, Cluzet V, Cordisco ME, Greenblatt B, Petrini J, Rausch W, Shade D, Lane K, Gawad AL, Klein SL, Pekosz A, Shoham S, Casadevall A, Bloch EM, Hanley D, Sullivan DJ, Tobian AAR. Dynamics of inflammatory responses after SARS-CoV-2 infection by vaccination status in the USA: a prospective cohort study. THE LANCET. MICROBE 2023; 4:e692-e703. [PMID: 37659419 PMCID: PMC10475695 DOI: 10.1016/s2666-5247(23)00171-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 09/04/2023]
Abstract
BACKGROUND Cytokines and chemokines play a critical role in the response to infection and vaccination. We aimed to assess the longitudinal association of COVID-19 vaccination with cytokine and chemokine concentrations and trajectories among people with SARS-CoV-2 infection. METHODS In this longitudinal, prospective cohort study, blood samples were used from participants enrolled in a multi-centre randomised trial assessing the efficacy of convalescent plasma therapy for ambulatory COVID-19. The trial was conducted in 23 outpatient sites in the USA. In this study, participants (aged ≥18 years) were restricted to those with COVID-19 before vaccination or with breakthrough infections who had blood samples and symptom data collected at screening (pre-transfusion), day 14, and day 90 visits. Associations between COVID-19 vaccination status and concentrations of 21 cytokines and chemokines (measured using multiplexed sandwich immunoassays) were examined using multivariate linear mixed-effects regression models, adjusted for age, sex, BMI, hypertension, diabetes, trial group, and COVID-19 waves (pre-alpha or alpha and delta). FINDINGS Between June 29, 2020, and Sept 30, 2021, 882 participants recently infected with SARS-CoV-2 were enrolled, of whom 506 (57%) were female and 376 (43%) were male. 688 (78%) of 882 participants were unvaccinated, 55 (6%) were partly vaccinated, and 139 (16%) were fully vaccinated at baseline. After adjusting for confounders, geometric mean concentrations of interleukin (IL)-2RA, IL-7, IL-8, IL-15, IL-29 (interferon-λ), inducible protein-10, monocyte chemoattractant protein-1, and tumour necrosis factor-α were significantly lower among the fully vaccinated group than in the unvaccinated group at screening. On day 90, fully vaccinated participants had approximately 20% lower geometric mean concentrations of IL-7, IL-8, and vascular endothelial growth factor-A than unvaccinated participants. Cytokine and chemokine concentrations decreased over time in the fully and partly vaccinated groups and unvaccinated group. Log10 cytokine and chemokine concentrations decreased faster among participants in the unvaccinated group than in other groups, but their geometric mean concentrations were generally higher than fully vaccinated participants at 90 days. Days since full vaccination and type of vaccine received were not correlated with cytokine and chemokine concentrations. INTERPRETATION Initially and during recovery from symptomatic COVID-19, fully vaccinated participants had lower concentrations of inflammatory markers than unvaccinated participants suggesting vaccination is associated with short-term and long-term reduction in inflammation, which could in part explain the reduced disease severity and mortality in vaccinated individuals. FUNDING US Department of Defense, National Institutes of Health, Bloomberg Philanthropies, State of Maryland, Mental Wellness Foundation, Moriah Fund, Octapharma, HealthNetwork Foundation, and the Shear Family Foundation.
Collapse
Affiliation(s)
- Xianming Zhu
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kelly A Gebo
- Department of Medicine, Division of Infectious Diseases, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Alison G Abraham
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Feben Habtehyimer
- Department of Medicine, Division of Infectious Diseases, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Eshan U Patel
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Oliver Laeyendecker
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Washington, DC, USA
| | - Thomas J Gniadek
- Department of Pathology and Laboratory Medicine, Northshore University Health System, Evanston, IL, USA
| | - Reinaldo E Fernandez
- Department of Medicine, Division of Infectious Diseases, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Owen R Baker
- Department of Medicine, Division of Infectious Diseases, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Malathi Ram
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Edward R Cachay
- Department of Medicine, Division of Infectious Diseases, University of California, San Diego, San Diego, CA, USA
| | - Judith S Currier
- Department of Medicine, Division of Infectious Diseases, University of California, Los Angeles, CA, USA
| | - Yuriko Fukuta
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan M Gerber
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sonya L Heath
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Barry Meisenberg
- Department of Medicine and Research Institute of Luminis Health, Annapolis, MD, USA
| | - Moises A Huaman
- Department of Medicine, Division of Infectious Diseases University of Cincinnati, Cincinnati, OH, USA
| | - Adam C Levine
- Department of Emergency Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Aarthi Shenoy
- Division of Hematology, Medstar DC Hospital, Washington, DC, USA
| | - Shweta Anjan
- Department of Medicine, Division of Infectious Diseases, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Janis E Blair
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic Hospital, Phoenix, AZ, USA
| | - Daniel Cruser
- Department of Pathology, Nuvance Health Vassar Brothers Medical Center, Poughkeepsie, NY, USA
| | - Donald N Forthal
- Department of Medicine, Division of Infectious Diseases, University of California, Irvine, CA, USA
| | - Laura L Hammitt
- Department of International Health, Johns Hopkins University, Baltimore, MD, USA
| | - Seble Kassaye
- Division of Infectious Diseases, Georgetown University Medical Center, Washington, DC, USA
| | - Giselle S Mosnaim
- Division of Allergy and Immunology, Department of Medicine, Northshore University Health System, Evanston, IL, USA
| | - Bela Patel
- Department of Medicine, Divisions of Pulmonary and Critical Care Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - James H Paxton
- Department of Emergency Medicine, Wayne State University, Detroit, MI, USA
| | - Jay S Raval
- Department of Pathology, University of New Mexico, Albuquerque, NM, USA
| | | | | | - Patrick Broderick
- Department of Emergency Medicine, Nuvance Health Danbury Hospital, Danbury, CT, USA
| | - Valerie Cluzet
- Department of Infectious Disease, Nuvance Health Vassar Brothers Medical Center, Poughkeepsie, NY, USA
| | - Marie Elena Cordisco
- Department of Emergency Medicine, Nuvance Health Danbury Hospital, Danbury, CT, USA
| | - Benjamin Greenblatt
- Department of Emergency Medicine, Nuvance Health Norwalk Hospital, Norwark, CT, USA
| | - Joann Petrini
- Department of Emergency Medicine, Nuvance Health Danbury Hospital, Danbury, CT, USA
| | - William Rausch
- Department of Emergency Medicine, Nuvance Health Danbury Hospital, Danbury, CT, USA
| | - David Shade
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Karen Lane
- Department of Neurology, Brain Injury Outcomes Division, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Amy L Gawad
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - Sabra L Klein
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew Pekosz
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - Evan M Bloch
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Daniel Hanley
- Department of Neurology, Brain Injury Outcomes Division, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - David J Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - Aaron A R Tobian
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
355
|
Ho JSY, Sia CH, Ngiam JN, Loh PH, Chew NWS, Kong WKF, Poh KK. A review of COVID-19 vaccination and the reported cardiac manifestations. Singapore Med J 2023; 64:543-549. [PMID: 34808708 PMCID: PMC10564100 DOI: 10.11622/smedj.2021210] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022]
Abstract
In Singapore, 9.03 million doses of the mRNA COVID-19 vaccines by Pfizer-BioNTech and Moderna have been administered, and 4.46 million people are fully vaccinated. An additional 87,000 people have been vaccinated with vaccines in World Health Organization's Emergency Use Listing. The aim of this review is to explore the reported cardiac adverse events associated with different types of COVID-19 vaccines. A total of 42 studies that reported cardiac side effects after COVID-19 vaccination were included in this study. Reported COVID-19 vaccine-associated cardiac adverse events were mainly myocarditis and pericarditis, most commonly seen in adolescent and young adult male individuals after mRNA vaccination. Reports of other events such as acute myocardial infarction, arrhythmia and stress cardiomyopathy were rare. Outcomes of post-vaccine myocarditis and pericarditis were good. Given the good vaccine efficacy and the high number of cases of infection, hospitalisation and death that could potentially be prevented, COVID-19 vaccine remains of overall benefit, based on the current available data.
Collapse
Affiliation(s)
- Jamie Sin Ying Ho
- Academic Foundation Programme, Royal Free London NHS Trust, London, UK
| | - Ching-Hui Sia
- Department of Cardiology, National University Heart Centre Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jinghao Nicholas Ngiam
- Division of Infectious Diseases, Department of Medicine, National University Health System, Singapore
| | - Poay Huan Loh
- Department of Cardiology, National University Heart Centre Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - William Kok-Fai Kong
- Department of Cardiology, National University Heart Centre Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kian-Keong Poh
- Department of Cardiology, National University Heart Centre Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
356
|
Labani A, Chou S, Kaviani K, Ropero B, Russman K, Becker D. Incidence of multiple sclerosis relapses and pseudo-relapses following COVID-19 vaccination. Mult Scler Relat Disord 2023; 77:104865. [PMID: 37418929 DOI: 10.1016/j.msard.2023.104865] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/13/2023] [Accepted: 06/29/2023] [Indexed: 07/09/2023]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic created an urgency for an effective vaccine. The FDA approved vaccines offered by Pfizer-BioNTech (BNT162b2), ModernaTX (mRNA-1273) and Janssen/Johnson & Johnson (Ad26.COV2.S) have shown minimal side effects (SE) in general population studies. Multiple sclerosis (MS) patients were not specifically represented in the above studies. The MS community is interested in how these vaccines behave in people with MS. In this study, we compare the SE experienced by MS to that of the general population after SARS-CoV-2 vaccination and evaluate their risk of relapses or pseudo-relapses. METHODS A retrospective, single-site, cohort study of 250 MS patients who received the initial cycle of FDA approved SARS-CoV-2 vaccines with 151 of whom also received an additional booster dose. SE resulting immediately after COVID-19 vaccination were collected as part of the standard clinical care during patient visits. RESULTS Out of the studied 250 MS patients, 135 received the first and second doses of BNT162b2 with less than 1% and 4% pseudo-relapses respectively and 79 received the third BNT162b2 dose with a pseudo-relapse rate of 3%. 88 received the mRNA-1273 vaccine with a pseudo-relapse frequency of 2% and 5% after the first and second doses respectively. 70 patients had the mRNA-1273 vaccine booster with a 3% pseudo-relapse rate. 27 received the Ad26.COV2.S first dose, 2 of whom received a second Ad26.COV2.S booster dose, with no reports of MS worsening. No acute relapses were reported in our patient population. All patients experiencing pseudo-relapse symptoms returned to baseline within 96 h. CONCLUSION COVID-19 vaccine is safe in patients with MS. Cases of temporary worsening of MS symptoms following SARS-CoV-2 are rare. Our findings support those reported by other recent studies and the CDC recommendation for MS patients to receive the FDA-approved COVID-19 vaccines, including the boosters.
Collapse
Affiliation(s)
- Amir Labani
- International Neurorehabilitation Institute, Lutherville, MD, United States
| | - Scott Chou
- International Neurorehabilitation Institute, Lutherville, MD, United States
| | - Kasra Kaviani
- International Neurorehabilitation Institute, Lutherville, MD, United States
| | - Brenda Ropero
- International Neurorehabilitation Institute, Lutherville, MD, United States
| | - Katharine Russman
- International Neurorehabilitation Institute, Lutherville, MD, United States
| | - Daniel Becker
- International Neurorehabilitation Institute, Lutherville, MD, United States; John Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, United States.
| |
Collapse
|
357
|
Bassetti M, Brucci G, Vena A, Giacobbe DR. Use of antibiotics in hospitalized patients with COVID-19: evolving concepts in a highly dynamic antimicrobial stewardship scenario. Expert Opin Pharmacother 2023; 24:1679-1684. [PMID: 37466425 DOI: 10.1080/14656566.2023.2239154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/23/2023] [Accepted: 07/18/2023] [Indexed: 07/20/2023]
Abstract
INTRODUCTION Excessive use of antibiotics has been frequently reported in hospitalized patients with COVID-19 worldwide, compared to the actual number of bacterial co-infections or super-infections. AREAS COVERED In this perspective, we discuss the current literature on the use of antibiotics and antimicrobial stewardship interventions in hospitalized patients with COVID-19. A search was conducted in PubMed up to March 2023. EXPERT OPINION The COVID-19 pandemic has witnessed an excessive use of antibiotics in hospitals worldwide, especially before the advent of COVID-19 vaccination, although according to the most recent data there is still an important disproportion between the prevalence of antibiotic use and that of proven bacterial coinfection or superinfections. An important reduction in the prevalence of antibiotic use in COVID-19 patients reported in the literature, from 70-100% to 50-60%, has been observed after successful vaccination campaigns, likely related to the reduced median disease severity of hospitalized COVID-19 patients and some successful interventions of antimicrobial and diagnostic stewardship. However, the disproportion between antibiotic use and the prevalence of bacterial infections (4-6%) is still uncomfortable from an antimicrobial stewardship perspective and requires further attention.
Collapse
Affiliation(s)
- Matteo Bassetti
- Infectious Diseases Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Giorgia Brucci
- Infectious Diseases Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Antonio Vena
- Infectious Diseases Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Daniele Roberto Giacobbe
- Infectious Diseases Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| |
Collapse
|
358
|
van der Arend BWH, Bloemhof MM, van der Schoor AG, van Zwet EW, Terwindt GM. Effect of COVID vaccination on monthly migraine days: a longitudinal cohort study. Cephalalgia 2023; 43:3331024231198792. [PMID: 37684012 DOI: 10.1177/03331024231198792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
BACKGROUND This longitudinal cohort study aimed to investigate changes in migraine-related outcomes following COVID-19 infection and vaccination. METHODS We identified 547 clinically diagnosed migraine patients from the Leiden Headache Center who kept a headache E-diary during the COVID-19 pandemic (February 2020 to August 2022). We sent a questionnaire to register their COVID-19 infection and/or vaccination dates. After applying inclusion criteria, n = 59 participants could be included in the infection analysis and n = 147 could be included in the vaccination analysis. Primary outcome was the change in monthly migraine days (MMD) between 1 month prior and 1 month post COVID-19 infection or vaccination. Secondary outcome variables were change in monthly headache days (MHD) and monthly acute medication days (MAMD). RESULTS Vaccination against COVID-19 was associated with an increase in MMD (1.06; 95% confidence interval [CI] = 0.57-1.55; p < 0.001), MHD (1.52; 95% CI = 0.91-2.14; p < 0.001) and MAMD (0.72; 95% CI = 0.33-1.12; p < 0.001) in the first month post-vaccination. COVID-19 infection solely increased the number of MAMD (1.11; 95% CI = 0.10-1.62; p < 0.027), but no statistically significant differences in MMD or MHD were observed. CONCLUSIONS Our findings imply that vaccination against COVID-19 is associated with an increase in migraine, indicating a possible role of inflammatory mediators in migraine pathophysiology.
Collapse
Affiliation(s)
- Britt W H van der Arend
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Internal Medicine, Erasmus University, Rotterdam, The Netherlands
| | - Mirthe M Bloemhof
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Erik W van Zwet
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
359
|
Wang J, Ai J, Xiang H, Zhang Y, Hou Z, Zhang Q, Lv J, Chen S, Liu C, Li Q, Liang J, Xie F, Jiang S, Zhang N, Zhang A, Lan X, Zhang X, Li J, Liu D, Wang W, Rao W, Qun Z, Tian Q, Qi X, Zhang W. Immunogenicity and safety of a booster COVID‐19 vaccination in patients with chronic liver disease: A multicenter study. PORTAL HYPERTENSION & CIRRHOSIS 2023; 2:127-135. [DOI: 10.1002/poh2.57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 08/06/2023] [Indexed: 01/03/2025]
Abstract
AbstractAimPatients with chronic liver disease (CLD), especially cirrhosis, are at a high risk of severe illness or death from coronavirus disease‐2019 (COVID‐19) and may have a suboptimal immune response to the severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) vaccine. This study aimed to evaluate the safety and immunogenicity of the COVID‐19 booster vaccination in patients with CLD.MethodsThe study protocol was prospectively registered at ClinicalTrials.gov (No. NCT05204602) after approval by the Ethics Committee. Adult participants with CLD were enrolled in this multicenter prospective study. They completed two doses of the inactivated COVID‐19 vaccine and received booster doses at least 6 months later. Adverse reactions were recorded within 14 days after the booster dose. Serum samples of the enrolled patients were collected before and after booster vaccination and tested for SARS‐CoV‐2 receptor‐binding domain (RBD) immunoglobulin G and neutralizing antibodies. The chi‐squared or Fisher's exact test was used to compare categorical data, and the Mann–Whitney U test was used to compare continuous variables. Two‐sided p < 0.05 were considered statistically significant.ResultsIn total, 63 patients were enrolled from four hospitals in China, including 29 patients with cirrhosis. The median age of all patients was 55 years, and 61.9% (39/63) were male. The vaccines were well tolerated; most adverse reactions were mild and transient, and injection site pain (6.4%; 4/63) and fatigue (3.2%, 2/63) were the most frequent local and systemic adverse events. Following the booster vaccination, our results showed that in the whole cohort, the levels and positive rates of anti‐RBD IgG and neutralizing antibodies were significantly higher than baseline levels (all p < 0.05).ConclusionsThe inactivated COVID‐19 booster vaccine was safe and significantly increased antibody levels and positivity rates following standard vaccination regimens in patients with CLD, especially those with cirrhosis.
Collapse
Affiliation(s)
- Jitao Wang
- Department of Radiology, Center of Portal Hypertension, Zhongda Hospital, School of Medicine Southeast University Nanjing Jiangsu China
- Xingtai Key Laboratory of Precision Medicine for Liver Cirrhosis and Portal Hypertension Xingtai People's Hospital of Hebei Medical University Xingtai Hebei China
| | - Jingwen Ai
- Department of Infectious Diseases Huashan Hospital Affiliated to Fudan University Shanghai China
| | - Huiling Xiang
- Department of Hepatology and Gastroenterology The Third Central Hospital of Tianjin Tianjin China
| | - Yanliang Zhang
- Department of Infectious Diseases Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine Nanjing Jiangsu China
| | - Zhiyun Hou
- Department of Hepatobiliary Surgery Jincheng People's Hospital Jincheng Shanxi China
| | - Qiran Zhang
- Department of Infectious Diseases Huashan Hospital Affiliated to Fudan University Shanghai China
| | - Jiaojian Lv
- Department of Infectious Diseases Lishui People's Hospital Lishui Zhejiang China
| | - Shubo Chen
- Xingtai Key Laboratory of Precision Medicine for Liver Cirrhosis and Portal Hypertension Xingtai People's Hospital of Hebei Medical University Xingtai Hebei China
| | - Chuan Liu
- Department of Radiology, Center of Portal Hypertension, Zhongda Hospital, School of Medicine Southeast University Nanjing Jiangsu China
| | - Qianqian Li
- Department of Hepatology and Gastroenterology The Third Central Hospital of Tianjin Tianjin China
| | - Jing Liang
- Department of Hepatology and Gastroenterology The Third Central Hospital of Tianjin Tianjin China
| | - Faren Xie
- Department of Infectious Diseases Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine Nanjing Jiangsu China
| | - Shujun Jiang
- Department of Infectious Diseases Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine Nanjing Jiangsu China
| | - Nina Zhang
- Department of Hepatobiliary Surgery Jincheng People's Hospital Jincheng Shanxi China
| | - Aiguo Zhang
- Department of Hepatobiliary Surgery Jincheng People's Hospital Jincheng Shanxi China
| | - Xiaolin Lan
- Department of Infectious Diseases Lishui People's Hospital Lishui Zhejiang China
| | - Xuying Zhang
- Clinal Laboratory Lishui People's Hospital Lishui Zhejiang China
| | - Jinlong Li
- Xingtai Key Laboratory of Precision Medicine for Liver Cirrhosis and Portal Hypertension Xingtai People's Hospital of Hebei Medical University Xingtai Hebei China
| | - Dengxiang Liu
- Xingtai Key Laboratory of Precision Medicine for Liver Cirrhosis and Portal Hypertension Xingtai People's Hospital of Hebei Medical University Xingtai Hebei China
| | - Wenchuan Wang
- Xingtai Key Laboratory of Precision Medicine for Liver Cirrhosis and Portal Hypertension Xingtai People's Hospital of Hebei Medical University Xingtai Hebei China
| | - Wei Rao
- Liver Disease Center, Organ Transplant Center Affiliated Hospital of Qingdao University Qingdao Shandong China
| | - Zhang Qun
- Liver Disease Center, Organ Transplant Center Affiliated Hospital of Qingdao University Qingdao Shandong China
| | - Qiuju Tian
- Liver Disease Center, Organ Transplant Center Affiliated Hospital of Qingdao University Qingdao Shandong China
| | - Xiaolong Qi
- Department of Radiology, Center of Portal Hypertension, Zhongda Hospital, School of Medicine Southeast University Nanjing Jiangsu China
| | - Wenhong Zhang
- Department of Infectious Diseases Huashan Hospital Affiliated to Fudan University Shanghai China
| |
Collapse
|
360
|
Cabral G, Azurara L, Ferreira JC. Relapse of Kleine-Levin Syndrome From COVID-19 Vaccine: Causal or Coincidence? J Clin Neurol 2023; 19:512-513. [PMID: 37635431 PMCID: PMC10471551 DOI: 10.3988/jcn.2023.0132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 08/29/2023] Open
Affiliation(s)
- Gonçalo Cabral
- Department of Neurology, Hospital Egas Moniz, Centro Hospitalar Lisboa Ocidental, Lisboa, Portugal.
| | - Laura Azurara
- Department of Child Neurology, Hospital São Francisco Xavier, Centro Hospitalar Lisboa Ocidental, Lisboa, Portugal
| | - José Carlos Ferreira
- Department of Child Neurology, Hospital São Francisco Xavier, Centro Hospitalar Lisboa Ocidental, Lisboa, Portugal
| |
Collapse
|
361
|
Medina J, Rojas-Cessa R, Dong Z, Umpaichitra V. A global blockchain for recording high rates of COVID-19 vaccinations. Comput Biol Med 2023; 163:107074. [PMID: 37311384 PMCID: PMC10228165 DOI: 10.1016/j.compbiomed.2023.107074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 05/13/2023] [Accepted: 05/27/2023] [Indexed: 06/15/2023]
Abstract
Blockchain has been recently proposed to securely record vaccinations against COVID-19 and manage their verification. However, existing solutions may not fully meet the requirements of a global vaccination management system. These requirements include the scalability required to support a global vaccination campaign, like one against COVID-19, and the capability to facilitate the interoperation between the independent health administrations of different countries. Moreover, access to global statistics can help to control securing community health and provide continuity of care for individuals during a pandemic. In this paper, we propose GEOS, a blockchain-based vaccination management system designed to address the challenges faced by the global vaccination campaign against COVID-19. GEOS offers interoperability between vaccination information systems at both domestic and international levels, supporting high vaccination rates and extensive coverage for the global population. To provide those features, GEOS uses a two-layer blockchain architecture, a simplified byzantine-tolerant consensus algorithm, and the Boneh-Lynn-Shacham signature scheme. We analyze the scalability of GEOS by examining transaction rate and confirmation times, considering factors such as the number of validators, communication overhead, and block size within the blockchain network. Our findings demonstrate the effectiveness of GEOS in managing COVID-19 vaccination records and statistical data for 236 countries, encompassing crucial information such as daily vaccination rates for highly populous nations and the global vaccination demand, as identified by the World Health Organization.
Collapse
Affiliation(s)
- Jorge Medina
- New Jersey Institute of Technology, Department of Electrical and Computer Engineering, Newark, NJ, 07102, USA.
| | - Roberto Rojas-Cessa
- New Jersey Institute of Technology, Department of Electrical and Computer Engineering, Newark, NJ, 07102, USA.
| | - Ziqian Dong
- New York Institute of Technology, Department of Electrical and Computer Engineering, New York, NY, 10023, USA.
| | | |
Collapse
|
362
|
Massy N, Atzenhoffer M, Boulay C, Pecquet PE, Ledys F, Cracowski JL, Masmoudi K, Lepelley M, Gras-Champel V. [COVID-19 and adenovirus vaccines: French experience of enhanced pharmacovigilance]. Therapie 2023; 78:489-498. [PMID: 36759287 PMCID: PMC9862661 DOI: 10.1016/j.therap.2023.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
As part of the COVID-19 vaccination campaign, the National Agency for the Safety of Medicines and Health Products and all 31 regional pharmacovigilance centers were mobilized in an exceptional reinforced vaccine pharmacovigilance surveillance system. Concerning adenovirus vaccines, Vaxzévria® and Jcovden®, this national system, based on the daily analysis of notified cases of adverse events, has allowed the early identification of safety signals, some of which have been validated, others still under analysis, common to mRNA vaccines or more specific of adenovirus vaccines such as Vaccine Induced Immune Thrombocytopenia. Complementing european and international actions, this follow-up has contributed to a better definition of the safety profile of these vaccines and has led to redefine the vaccine strategy in our country. Although today these two vaccines have no longer place in the national vaccine strategy, they are still used in other countries, where the experience acquired could be useful and will contribute to fuel the reflection on future therapies involving viral vectors.
Collapse
Affiliation(s)
- Nathalie Massy
- Centre régional de pharmacovigilance, service de pharmacologie, CHU Rouen, 76031 Rouen, France
| | - Marina Atzenhoffer
- Centre régional de pharmacovigilance, service hospitalo-universitaire de pharmacologie et toxicologie, hospices civils de Lyon, 69424 Lyon, France
| | - Charlène Boulay
- Centre régional de pharmacovigilance, service de pharmacologie, CHU Rouen, 76031 Rouen, France
| | - Pauline-Eva Pecquet
- Centre régional de pharmacovigilance, service de pharmacologie clinique, CHU Amiens-Picardie, 80054 Amiens, France
| | - Fanny Ledys
- Centre régional de pharmacovigilance, service hospitalo-universitaire de pharmacologie et toxicologie, hospices civils de Lyon, 69424 Lyon, France
| | - Jean-Luc Cracowski
- Centre régional de pharmacovigilance, service de pharmacologie, CHU Grenoble, 38043 Grenoble cedex 09, France
| | - Kamel Masmoudi
- Centre régional de pharmacovigilance, service de pharmacologie clinique, CHU Amiens-Picardie, 80054 Amiens, France
| | - Marion Lepelley
- Centre régional de pharmacovigilance, service de pharmacologie, CHU Grenoble, 38043 Grenoble cedex 09, France
| | - Valérie Gras-Champel
- Centre régional de pharmacovigilance, service de pharmacologie clinique, CHU Amiens-Picardie, 80054 Amiens, France.
| |
Collapse
|
363
|
Morel J. Infection prevention and vaccination in the rheumatic diseases. Joint Bone Spine 2023; 90:105568. [PMID: 36990142 DOI: 10.1016/j.jbspin.2023.105568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2022] [Indexed: 03/29/2023]
Abstract
Patients with chronic inflammatory rheumatisms (CIR) have a higher risk of infections compared to healthy subjects. Viral and bacterial pneumonia are the most frequent infections observed in CIR with targeted disease modifying anti-rheumatic drugs (DMARDs). Moreover, drugs used to treat CIR (especially biologic and synthetic targeted DMARDs) increase the risk of infection and expose CIR patients to opportunistic infections such as tuberculosis reactivation. To limit the risk of infection, the risk-benefit ratio should be evaluated for each patient based on their characteristics and comorbidities. To prevent infections, an initial pre-treatment work-up must be performed, especially before the initiation of conventional synthetic DMARDs or biological and synthetic targeted DMARDs. This pre-treatment assessment includes the case history, laboratory and radiology findings as well. The physician must make sure a patient's vaccinations are up-to-date. The vaccines recommended for patients with CIR being treated with conventional synthetic DMARDs, bDMARDs, tsDMARDs and/or steroids should be given. Patient education is also very important. During workshops, they learn how to manage their drug treatments in at-risk situations and learn which symptoms require treatment discontinuation.
Collapse
Affiliation(s)
- Jacques Morel
- Rheumatology Department, CHU and University of Montpellier, PhyMedExp, Inserm, CNRS, Montpellier, France.
| |
Collapse
|
364
|
Jeong HS, Chun BC. Signal detection of COVID-19 vaccines adverse events using spontaneous reports from South Korea. Pharmacoepidemiol Drug Saf 2023; 32:961-968. [PMID: 37019851 DOI: 10.1002/pds.5629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/14/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023]
Abstract
PURPOSE Studies on the detection of COVID-19 vaccine signals in South Korea are insufficient. Therefore, to investigate adverse events (AEs) that might be associated with COVID-19 vaccines, signals were detected using spontaneous reports from South Korea. We compared the signals with the vaccine insert lists of the regulators in the four countries. METHODS Spontaneous reports from 62 sites were collected by the National Medical Center between January 2013 and May 2022. A descriptive analysis of AEs associated with COVID-19 vaccines (Pfizer, Moderna, AstraZeneca, and Janssen) was performed, and the proportional reporting ratio, reporting odds ratio, and information component were calculated. We performed five analyses, with five cases and one control group. RESULTS During the study period, 68 355 cases were reported, of which 12 485 were COVID-19 vaccine AEs. Injection site pain (2198 cases, 17.6%), myalgia (1552 cases, 12.4%), headache (1145 cases, 9.2%), pyrexia (1003 cases, 8.0%), and fatigue (735 cases, 5.9%) were frequently reported. When comparing all COVID-19 vaccines with other viral vaccines, 20 signals were detected, of which cachexia, dyspepsia, abdominal discomfort, and mood swings were not listed on the vaccine inserts in all four countries. Overall, 20, 17, 29, and 9 signals were detected in vaccines developed by Pfizer, Moderna, AstraZeneca, and Janssen, respectively. CONCLUSIONS Based on a disproportionate analysis of COVID-19 vaccine AEs using spontaneous reports from South Korea, different signals were detected for each vaccine manufacturer.
Collapse
Affiliation(s)
- Hye Su Jeong
- Drug Safety Monitoring Center, National Medical Center, Seoul, South Korea
- Department of Epidemiology and Health Informatics, Graduate School of Public Health, Korea University, Seoul, South Korea
| | - Byung Chul Chun
- Department of Epidemiology and Health Informatics, Graduate School of Public Health, Korea University, Seoul, South Korea
- Department of Preventive Medicine, Korea University College of Medicine, Seoul, South Korea
| |
Collapse
|
365
|
Tokunoh N, Tamiya S, Watanabe M, Okamoto T, Anindita J, Tanaka H, Ono C, Hirai T, Akita H, Matsuura Y, Yoshioka Y. A nasal vaccine with inactivated whole-virion elicits protective mucosal immunity against SARS-CoV-2 in mice. Front Immunol 2023; 14:1224634. [PMID: 37720231 PMCID: PMC10500122 DOI: 10.3389/fimmu.2023.1224634] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/14/2023] [Indexed: 09/19/2023] Open
Abstract
Introduction Vaccinations are ideal for reducing the severity of clinical manifestations and secondary complications of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); however, SARS-CoV-2 continues to cause morbidity and mortality worldwide. In contrast to parenteral vaccines such as messenger RNA vaccines, nasal vaccines are expected to be more effective in preventing viral infections in the upper respiratory tract, the primary locus for viral infection and transmission. In this study, we examined the prospects of an inactivated whole-virion (WV) vaccine administered intranasally against SARS-CoV-2. Methods Mice were immunized subcutaneously (subcutaneous vaccine) or intranasally (nasal vaccine) with the inactivated WV of SARS-CoV-2 as the antigen. Results The spike protein (S)-specific IgA level was found to be higher upon nasal vaccination than after subcutaneous vaccination. The level of S-specific IgG in the serum was also increased by the nasal vaccine, although it was lower than that induced by the subcutaneous vaccine. The nasal vaccine exhibited a stronger defense against viral invasion in the upper respiratory tract than the subcutaneous vaccine and unimmunized control; however, both subcutaneous and nasal vaccines provided protection in the lower respiratory tract. Furthermore, we found that intranasally administered inactivated WV elicited robust production of S-specific IgA in the nasal mucosa and IgG in the blood of mice previously vaccinated with messenger RNA encoding the S protein. Discussion Overall, these results suggest that a nasal vaccine containing inactivated WV can be a highly effective means of protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Nagisa Tokunoh
- Innovative Vaccine Research and Development Center, The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Shigeyuki Tamiya
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Department of Microbiology and Immunology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Masato Watanabe
- Innovative Vaccine Research and Development Center, The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
| | - Toru Okamoto
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Jessica Anindita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Science, Chiba University, Chiba-shi, Chiba, Japan
| | - Hiroki Tanaka
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Chikako Ono
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Toshiro Hirai
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita, Osaka, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Yoshiharu Matsuura
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita, Osaka, Japan
| | - Yasuo Yoshioka
- Innovative Vaccine Research and Development Center, The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita, Osaka, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
366
|
Bostanghadiri N, Ziaeefar P, Mofrad MG, Yousefzadeh P, Hashemi A, Darban-Sarokhalil D. COVID-19: An Overview of SARS-CoV-2 Variants-The Current Vaccines and Drug Development. BIOMED RESEARCH INTERNATIONAL 2023; 2023:1879554. [PMID: 37674935 PMCID: PMC10480030 DOI: 10.1155/2023/1879554] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/07/2023] [Accepted: 08/04/2023] [Indexed: 09/08/2023]
Abstract
The world is presently in crisis facing an outbreak of a health-threatening microorganism known as COVID-19, responsible for causing uncommon viral pneumonia in humans. The virus was first reported in Wuhan, China, in early December 2019, and it quickly became a global concern due to the pandemic. Challenges in this regard have been compounded by the emergence of several variants such as B.1.1.7, B.1.351, P1, and B.1.617, which show an increase in transmission power and resistance to therapies and vaccines. Ongoing researches are focused on developing and manufacturing standard treatment strategies and effective vaccines to control the pandemic. Despite developing several vaccines such as Pfizer/BioNTech and Moderna approved by the U.S. Food and Drug Administration (FDA) and other vaccines in phase 4 clinical trials, preventive measures are mandatory to control the COVID-19 pandemic. In this review, based on the latest findings, we will discuss different types of drugs as therapeutic options and confirmed or developing vaccine candidates against SARS-CoV-2. We also discuss in detail the challenges posed by the variants and their effect on therapeutic and preventive interventions.
Collapse
Affiliation(s)
- Narjess Bostanghadiri
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Pardis Ziaeefar
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morvarid Golrokh Mofrad
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Parsa Yousefzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
367
|
Cosma C, Galla L, Padoan A, Furlan G, Marchioro L, Zaninotto M, Basso D, Plebani M. SARS-CoV-2 specific T-cell humoral response assessment after COVID-19 vaccination using a rapid direct real-time PCR amplification. Clin Chem Lab Med 2023; 61:1652-1660. [PMID: 36957995 DOI: 10.1515/cclm-2023-0129] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/01/2023] [Indexed: 03/25/2023]
Abstract
OBJECTIVES The SARS-CoV-2 immune response is mediated by both humoral and cellular immunity. In this study, SARS-CoV-2 specific cellular immunity was tested by a novel direct real-time PCR (dRT-PCR) assay, targeting mRNA of CXCL10, and compared with respect to an ELISA measuring interferon gamma (IFN-γ) release. METHODS Whole blood (Li-He) and serum samples were collected from 92 healthcare workers (HCW), with three doses of homologous (Pfizer/BioNTech, n=74) or heterologous (Pfizer/BioNTech and Vaxzevria or Moderna, n=18) vaccinations. Li-He samples were incubated with SCV2 PANEL-1-T-ACTIVATION (Hyris srl, Lodi, Italy), or CoV-2 IGRA TUBE ELISA (Euroimmune, Lubeck, Germany). CXCL10 mRNA expression was analyzed by bCube/bApp (Hyris), while IFN-γ was evaluated by quant-T-Cell SARS-CoV-2 ELISA (Euroimmune). Anti-SARS-CoV-2 S-RBD IgG levels were measured in sera using a CLIA assay (Snibe, Shenzen, China). RESULTS Imprecision of dRT-PCR assay was found to be satisfactory, and the two methods for measuring T cell immunity to SARS-CoV-2 peptides agreed in 82/87 (94.2%) of results. At qualitative dRT-PCR analyses, 81 subjects (93.2%) resulted as reactive to SARS-CoV-2 peptides, 3 (3.4%) were borderline and 3 were negative (3.4%). At univariate and multivariate analyses of quantitative dRT-PCR mRNA of CXCL10 and IFN-γ release results showed no difference between HCW with previous infection, homologous/heterologous vaccination, or demographical features. Anti-SARS-CoV-2 S-RBD IgG was associated with the previous infection and the time between the last vaccination or positivity. CONCLUSIONS Direct RT-PCR appeared accurate for determining the presence or absence of immunoreactivity of SARS-CoV-2 specific T cells, especially when rapid analyses are required, such as for organ transplantation.
Collapse
Affiliation(s)
- Chiara Cosma
- Laboratory Medicine Unit, University-Hospital of Padova, Padova, Italy
- QI.LAB.MED., Spin-off of the University of Padova, Padova, Italy
| | - Luisa Galla
- Laboratory Medicine Unit, University-Hospital of Padova, Padova, Italy
- QI.LAB.MED., Spin-off of the University of Padova, Padova, Italy
| | - Andrea Padoan
- Laboratory Medicine Unit, University-Hospital of Padova, Padova, Italy
- QI.LAB.MED., Spin-off of the University of Padova, Padova, Italy
- Department of Medicine-DIMED, University-Hospital of Padova, Padova, Italy
| | - Giulia Furlan
- QI.LAB.MED., Spin-off of the University of Padova, Padova, Italy
- Department of Medicine-DIMED, University-Hospital of Padova, Padova, Italy
| | - Lucio Marchioro
- QI.LAB.MED., Spin-off of the University of Padova, Padova, Italy
| | - Martina Zaninotto
- Laboratory Medicine Unit, University-Hospital of Padova, Padova, Italy
- QI.LAB.MED., Spin-off of the University of Padova, Padova, Italy
| | - Daniela Basso
- Laboratory Medicine Unit, University-Hospital of Padova, Padova, Italy
- QI.LAB.MED., Spin-off of the University of Padova, Padova, Italy
- Department of Medicine-DIMED, University-Hospital of Padova, Padova, Italy
| | - Mario Plebani
- QI.LAB.MED., Spin-off of the University of Padova, Padova, Italy
- Department of Medicine-DIMED, University-Hospital of Padova, Padova, Italy
| |
Collapse
|
368
|
Vollenberg R, Lorentzen EU, Kühn J, Nowacki TM, Meier JA, Trebicka J, Tepasse PR. Humoral Immunity in Immunosuppressed IBD Patients after the Third SARS-CoV-2 Vaccination: A Comparison with Healthy Control Subjects. Vaccines (Basel) 2023; 11:1411. [PMID: 37766088 PMCID: PMC10536352 DOI: 10.3390/vaccines11091411] [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: 07/24/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
INTRODUCTION The COVID-19 pandemic is a result of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Vaccination against COVID-19 is crucial for preventing severe illness and controlling the pandemic. This study aimed to examine how immunosuppressed patients with inflammatory bowel disease (IBD) responded to the third mRNA vaccination against SARS-CoV-2. The patients were undergoing treatments such as anti-TNF (infliximab, adalimumab), anti-α4ß7 integrin (vedolizumab), anti-IL12/23 (ustekinumab) and azathioprine (purine analog). Their responses were compared to those of healthy individuals. METHODS In this prospective study, 81 IBD patients and 15 healthy controls were enrolled 2-4 months after receiving the third mRNA vaccination. This study measured IgG antibody levels against the SARS-CoV-2 spike protein's receptor binding domain (RBD) and assessed potential neutralization capacity using a surrogate virus neutralization test (sVNT). RESULTS Overall, immunosuppressed IBD patients (without SARS-CoV-2 infection) exhibited significantly lower levels of anti-S-IgG (anti-RBD-IgG) and binding inhibition in the sVNT after the third vaccination compared to healthy controls. Patients under anti-TNF therapy showed notably reduced anti-S-IgG levels after the booster vaccination, in contrast to those receiving ustekinumab and azathioprine (p = 0.030, p = 0.031). IBD patients on anti-TNF therapy demonstrated significantly increased anti-S-IgG levels following prior SARS-CoV-2 infection (p = 0.020). CONCLUSION Even after the third vaccination, immunosuppressed IBD patients exhibited diminished humoral immunity compared to healthy controls, especially those on anti-TNF therapy. Cases of penetrating infections led to considerably higher antibody levels in IBD patients under anti-TNF therapy compared to uninfected patients. Further investigation through prospective studies in immunosuppressed IBD patients is needed to determine whether this effectively safeguards against future infections or severe disease.
Collapse
Affiliation(s)
- Richard Vollenberg
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology, University Hospital Muenster, 48149 Muenster, Germany; (J.T.); (P.-R.T.)
| | - Eva Ulla Lorentzen
- Institute of Virology, University Hospital Muenster, 48149 Muenster, Germany (J.K.)
| | - Joachim Kühn
- Institute of Virology, University Hospital Muenster, 48149 Muenster, Germany (J.K.)
| | - Tobias Max Nowacki
- Department of Medicine, Gastroenterology, Marienhospital Steinfurt, 48565 Steinfurt, Germany
| | - Jörn Arne Meier
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology, University Hospital Muenster, 48149 Muenster, Germany; (J.T.); (P.-R.T.)
| | - Jonel Trebicka
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology, University Hospital Muenster, 48149 Muenster, Germany; (J.T.); (P.-R.T.)
| | - Phil-Robin Tepasse
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology, University Hospital Muenster, 48149 Muenster, Germany; (J.T.); (P.-R.T.)
| |
Collapse
|
369
|
Ciarambino T, Crispino P, Buono P, Giordano V, Trama U, Iodice V, Leoncini L, Giordano M. Efficacy and Safety of Vaccinations in Geriatric Patients: A Literature Review. Vaccines (Basel) 2023; 11:1412. [PMID: 37766089 PMCID: PMC10537287 DOI: 10.3390/vaccines11091412] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
With the progressive lengthening of the average age of the population, especially in some countries such as Italy, vaccination of the elderly is a fixed point on which most of the public health efforts are concentrating as epidemic infectious diseases, especially those of the winter, have a major impact on the progression of severe disease, hospitalization, and death. The protection of the elderly against acute infectious diseases should not only limit mortality but also have a positive impact on the fragility of these people in terms of less disability and fewer care needs. However, vaccination of the elderly population differs in efficacy and safety compared to that of other population categories since aging and the consequent loss of efficiency of the immune system lead to a reduction in the immunogenicity of vaccines without achieving a lasting antibody coverage. There are various strategies to avoid the failure of immunization by vaccines such as resorting to supplementary doses with adjuvant vaccines, increasing the dosage of the antigen used, or choosing to inoculate the serum relying on various routes of administration of the vaccine. Vaccination in the elderly is also an important factor in light of growing antibiotic resistance because it can indirectly contribute to combating antibiotic resistance, reducing theoretically the use of those agents. Furthermore, vaccination in old age reduces mortality from infectious diseases preventable with vaccines and reduces the same rate of resistance to antibiotics. Given the importance and complexity of the topic, in this review, we will deal with the main aspects of vaccination in the elderly and how it can influence mortality and healthcare costs, especially in those countries where population aging is more evident. Therefore, we conducted a systematic literature search in PubMed to identify all types of studies published up to 31 May 2023 that examined the association between vaccination and the elderly. Data extraction and quality assessment were conducted by two reviewers (PC and TC) who independently extracted the following data and assessed the quality of each study.
Collapse
Affiliation(s)
- Tiziana Ciarambino
- Internal Medicine Department, Hospital of Marcianise, ASL Caserta, 81031 Caserta, Italy
- Direzione di Staff Direzione Generale Tutela per la Salute Regione Campania, 80143 Naples, Italy; (P.B.); (U.T.)
| | - Pietro Crispino
- Internal Medicine Department, Hospital of Latina, ASL Latina, 04100 Latina, Italy;
| | - Pietro Buono
- Direzione di Staff Direzione Generale Tutela per la Salute Regione Campania, 80143 Naples, Italy; (P.B.); (U.T.)
| | | | - Ugo Trama
- Direzione di Staff Direzione Generale Tutela per la Salute Regione Campania, 80143 Naples, Italy; (P.B.); (U.T.)
| | - Vincenzo Iodice
- ASL Caserta, Direttore Sanitario Aziendale, 81100 Caserta, Italy
| | - Laura Leoncini
- ASL Caserta, Direttore Sanitario, P.O. Marcianise, 81025 Marcianise, Italy
| | - Mauro Giordano
- Department of Advanced Medical and Surgical Science, University of Campania, L. Vanvitelli, 81100 Naples, Italy;
| |
Collapse
|
370
|
Schröder D, Müllenmeister C, Heinemann S, Hummers E, Klawonn F, Vahldiek K, Dopfer-Jablonka A, Steffens S, Mikuteit M, Niewolik J, Overbeck TR, Kallusky J, Königs G, Heesen G, Schmachtenberg T, Müller F. Social participation during the COVID-19 pandemic in persons with a high risk for a severe course of COVID-19 - results of a longitudinal, multi-center observational study in Germany. Health Psychol Behav Med 2023; 11:2249534. [PMID: 37645515 PMCID: PMC10461510 DOI: 10.1080/21642850.2023.2249534] [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: 02/28/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023] Open
Abstract
Objective The COVID-19 pandemic has affected how people go about their daily lives, often in various and substantial ways. This study aims to prospectively evaluate the changes in social participation during the COVID-19 pandemic in persons with a high risk for a severe COVID-19 course in Germany. Methods A paper-pencil-based survey was conducted starting at March 2021. Participants filled out questionnaires at four time points based on their COVID-19 vaccination status: before COVID-19 vaccination, one month, six months and twelve months after COVID-19 vaccination. Social participation measures included the Pandemic Social Participation Questionnaire (PSP-Q) and the Index for measuring participation restrictions (IMET). Repeated measures ANOVA and paired t-test were used to test for changes between time-points. Repeated measures correlation was used to assess the relationship between social participation and local COVID-19 incidences. Results Data from 245 participants was analyzed before and one month after COVID-19 vaccination. In addition, data from 156 participants was analyzed at time points one, six and twelve months after COVID-19. PSP-Q and IMET scores changed significantly after participants received a COVID-19 vaccination. Between one month and twelve months after vaccination, social participation improved significantly measured by PSP-Q. Social participation was negatively correlated with regional COVID-19 incidences before and after COVID-19 vaccination. Social participation was positively correlated with COVID-19 incidences between one month and twelve months after COVID-19 vaccination. Conclusions Social participation improved in persons with a high risk for a severe COVID-19 course during the pandemic. The local COVID-19 incidence showed a negative association with social participation only until the fall of 2021 when it was used as the sole metric to regulate COVID-19 protective measures. Although our data describes the trends in social participation, further studies are needed to identify the influencing factors for the observed increase in social participation.
Collapse
Affiliation(s)
- Dominik Schröder
- Department of General Practice, University Medical Center, Göttingen, Germany
| | | | - Stephanie Heinemann
- Department of General Practice, University Medical Center, Göttingen, Germany
- Department of Geriatrics, University Medical Center Göttingen, Göttingen, Germany
| | - Eva Hummers
- Department of General Practice, University Medical Center, Göttingen, Germany
| | - Frank Klawonn
- Department of Computer Science, Ostfalia University of Applied Sciences, Wolfenbuettel, Germany
- Biostatistics Group, Helmholtz Centre for Infecwetion Research, Braunschweig, Germany
| | - Kai Vahldiek
- Department of Computer Science, Ostfalia University of Applied Sciences, Wolfenbuettel, Germany
| | - Alexandra Dopfer-Jablonka
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner site Hannover-Braunschweig, Brunswick, Germany
| | - Sandra Steffens
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Marie Mikuteit
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Jacqueline Niewolik
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Tobias R. Overbeck
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Jonathan Kallusky
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Gloria Königs
- Department of General Practice, University Medical Center, Göttingen, Germany
| | - Gloria Heesen
- Department of General Practice, University Medical Center, Göttingen, Germany
| | - Tim Schmachtenberg
- Department of General Practice, University Medical Center, Göttingen, Germany
| | - Frank Müller
- Department of General Practice, University Medical Center, Göttingen, Germany
- Department of Family Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| |
Collapse
|
371
|
Iwahori K, Nii T, Yamaguchi N, Kawasaki T, Okamura S, Hashimoto K, Matsuki T, Tsujino K, Miki K, Osa A, Goya S, Abe K, Mori M, Takeda Y, Yamada T, Kida H, Kumanogoh A. A randomized phase 2 study on demeclocycline in patients with mild-to-moderate COVID-19. Sci Rep 2023; 13:13809. [PMID: 37612352 PMCID: PMC10447520 DOI: 10.1038/s41598-023-41051-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/21/2023] [Indexed: 08/25/2023] Open
Abstract
Tetracyclines exhibit anti-viral, anti-inflammatory, and immunomodulatory activities via various mechanisms. The present study investigated the efficacy and safety of demeclocycline in patients hospitalized with mild-to-moderate COVID-19 via an open-label, multicenter, parallel-group, randomized controlled phase 2 trial. Primary and secondary outcomes included changes from baseline (day 1, before the study treatment) in lymphocytes, cytokines, and SARS-CoV-2 RNA on day 8. Seven, seven, and six patients in the control, demeclocycline 150 mg daily, and demeclocycline 300 mg daily groups, respectively, were included in the modified intention-to-treat population that was followed until day 29. A significant change of 191.3/μL in the number of CD4+ T cells from day 1 to day 8 was observed in the demeclocycline 150 mg group (95% CI 5.1/μL-377.6/μL) (p = 0.023), whereas that in the control group was 47.8/μL (95% CI - 151.2/μL to 246.8/μL), which was not significant (p = 0.271). The change rates of CD4+ T cells negatively correlated with those of IL-6 in the demeclocycline-treated groups (R = - 0.807, p = 0.009). All treatment-emergent adverse events were of mild-to-moderate severity. The present results indicate that the treatment of mild-to-moderate COVID-19 patients with demeclocycline elicits immune responses conducive to recovery from COVID-19 with good tolerability.Trial registration: This study was registered with the Japan Registry of Clinical Trials (Trial registration number: jRCTs051200049; Date of the first registration: 26/08/2020).
Collapse
Affiliation(s)
- Kota Iwahori
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Takuro Nii
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka, Osaka, Japan
| | - Norihiko Yamaguchi
- Department of Respiratory Medicine, Kinki Central Hospital of Mutual Aid Association of Public School Teachers, Itami, Hyogo, Japan
| | - Takahiro Kawasaki
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Satomi Okamura
- Department of Medical Innovation, Osaka University Hospital, Suita, Osaka, Japan
| | - Kazuki Hashimoto
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka, Osaka, Japan
| | - Takanori Matsuki
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka, Osaka, Japan
| | - Kazuyuki Tsujino
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka, Osaka, Japan
| | - Keisuke Miki
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka, Osaka, Japan
| | - Akio Osa
- Department of Respiratory Medicine, Kinki Central Hospital of Mutual Aid Association of Public School Teachers, Itami, Hyogo, Japan
| | - Sho Goya
- Department of Respiratory Medicine, Kinki Central Hospital of Mutual Aid Association of Public School Teachers, Itami, Hyogo, Japan
| | - Kinya Abe
- Department of Internal Medicine, Toyonaka Municipal Hospital, Toyonaka, Osaka, Japan
| | - Masahide Mori
- Department of Thoracic Oncology, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka, Osaka, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tomomi Yamada
- Department of Medical Innovation, Osaka University Hospital, Suita, Osaka, Japan
| | - Hiroshi Kida
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Osaka, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Osaka, Japan
| |
Collapse
|
372
|
Struyf F, Hardt K, Van Rampelbergh R, Shukarev G, Inamdar A, Ruiz-Guiñazú J, van Paassen V, Anaya-Velarde L, Diba C, Ceuppens M, Cardenas V, Soff GA, Pragalos A, Sadoff J, Douoguih M. Thrombosis with thrombocytopenia syndrome: A database review of clinical trial and post-marketing experience with Ad26.COV2.S. Vaccine 2023; 41:5351-5359. [PMID: 37517912 DOI: 10.1016/j.vaccine.2023.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND Thrombosis with thrombocytopenia syndrome (TTS) is a very rare disorder described after vaccination with adenoviral vector-based COVID-19 vaccines. Co-occurring thrombosis with thrombocytopenia reported after vaccination can be a proxy for identification of TTS. METHODS Descriptive database review of all cases of co-occurring (within 42 days) thrombosis with thrombocytopenia in participants in Ad26.COV2.S clinical trials or recipients of Ad26.COV2.S in real-world clinical practice. Cases were retrieved from Janssens' clinical trial and Global Medical Safety databases. RESULTS There were 34 cases of co-occurring thrombosis with thrombocytopenia in Ad26.COV2.S recipients (46 per 100,000 person-years) and 15 after placebo (75 per 100,000 person-years) in clinical trials. Among Ad26.COV2.S recipients, mean age at the time of the event was 63 years (range 25-85), 82 % were male, mean time-to-onset 112 days (range 8-339) post-last Ad26.COV2.S dose, 26 events occurred post-dose-1, and 7 within a 28-day risk window post-vaccination. Diagnostic certainty was evaluated using Brighton Collaboration, US Centers for Disease Control and Prevention, and European Medicines Agency Pharmacovigilance Risk Assessment Committee case definitions. One case met the highest level of diagnostic certainty for all 3 definitions. There were 355 spontaneous reports of co-occurring thrombosis with thrombocytopenia in the Global Medical Safety database, 47 % males, 85 % within 28-days after vaccination. Twenty-seven cases met the highest level of diagnostic certainty for all definitions, 21 female, 19 with cerebral venous sinus thrombosis, age-range 18-68 years. Time-to-onset was 7-14 days post-vaccination in 20 cases. There were 8 fatalities. CONCLUSION TTS induced by Ad26.COV2.S is very rare. Most co-occurring thrombosis with thrombocytopenia does not constitute TTS.
Collapse
Affiliation(s)
- Frank Struyf
- Janssen Research and Development, Beerse, Belgium.
| | - Karin Hardt
- Janssen Research and Development, Beerse, Belgium
| | | | | | | | | | | | | | | | | | - Vicky Cardenas
- Janssen Research and Development, Spring House, PA, United States
| | - Gerald A Soff
- University of Miami Health System/Sylvester Comprehensive Cancer Center, Soffer Clinical Research Building, Miami, FL 33136, United States
| | | | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | | |
Collapse
|
373
|
Rosa RG, Falavigna M, Manfio JL, de Araujo CLP, Cohen M, do Valle Barbosa GRG, de Souza AP, Romeiro Silva FK, Sganzerla D, da Silva MMD, Ferreira D, de Oliveira Rodrigues C, de Souza EM, de Oliveira JC, Gradia DF, Brandalize APC, Royer CA, Luiz RM, Kucharski GA, Pedrotti F, Valluri SR, Srivastava A, Julião VW, Melone OC, Allen KE, Kyaw MH, Spinardi J, Del Carmen Morales Castillo G, McLaughlin JM. BNT162b2 mRNA COVID-19 against symptomatic Omicron infection following a mass vaccination campaign in southern Brazil: A prospective test-negative design study. Vaccine 2023; 41:5461-5468. [PMID: 37507274 DOI: 10.1016/j.vaccine.2023.07.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/01/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Evidence regarding effectiveness of BNT162b2 mRNA COVID-19 vaccine against Omicron in Latin America is limited. We estimated BNT162b2 effectiveness against symptomatic COVID-19 in Brazil when Omicron was predominant. METHODS This prospective test-negative, case-control study was conducted in Toledo, Brazil, following a mass COVID-19 vaccination with BNT162b2. Patients were included if they were aged ≥12 years, sought care for acute respiratory symptoms in the public health system between November 3, 2021 and June 20, 2022, and were tested for SARS-CoV-2 using RT-PCR. In the primary analysis, we determined the effectiveness of two doses of BNT162b2 against symptomatic COVID-19. RESULTS A total of 4,574 were enrolled; of these, 1,758 patients (586 cases and 1,172 controls) were included in the primary analysis. Mean age was 27.7 years, 53.8 % were women, and 90.1 % had a Charlson comorbidity index of zero. Omicron accounted for >97 % of all identified SARS-CoV-2 variants, with BA.1 and BA.2 accounting for 84.3 % and 12.6 %, respectively. Overall adjusted estimate of two-dose vaccine effectiveness against symptomatic COVID-19 was 46.7 % (95 %CI, 19.9 %-64.6 %) after a median time between the second dose and the beginning of COVID-19 symptoms of 94 days (IQR, 60-139 days). Effectiveness waned from 77.7 % at 7-29 days after receipt of a second dose to <30 % (non-significant) after ≥120 days. CONCLUSION In a relatively young and healthy Brazilian population, two doses of BNT162b2 provided protection against symptomatic Omicron infection. However, this protection waned significantly over time, underscoring the need for boosting with variant-adapted vaccines in this population prior to waves of disease activity. TRIAL REGISTRATION NUMBER ClinicalTrials.gov number, NCT05052307 (https://clinicaltrials.gov/ct2/show/NCT05052307).
Collapse
Affiliation(s)
- Regis Goulart Rosa
- Internal Medicine Department, Hospital Moinhos de Vento (HMV), Porto Alegre, RS, Brazil; Research Unit, Inova Medical, Porto Alegre, RS, Brazil; Research Institute, HMV, Porto Alegre, RS, Brazil.
| | - Maicon Falavigna
- Research Unit, Inova Medical, Porto Alegre, RS, Brazil; Research Institute, HMV, Porto Alegre, RS, Brazil; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | | | | | - Mírian Cohen
- Research Institute, HMV, Porto Alegre, RS, Brazil; Federal University of Rio Grande do Sul (UFRGS), Brazil
| | | | | | | | | | | | | | | | | | | | - Daniela Fiori Gradia
- Department of Biochemistry and Molecular Biology, Department of Genetics - UFPR, Brazil
| | | | - Carla Adriane Royer
- Department of Biochemistry and Molecular Biology, Department of Genetics - UFPR, Brazil
| | - Rafael Messias Luiz
- Faculty of Medicine - Campus Toledo - Federal University of Paraná (UFPR), Brazil
| | | | | | - Srinivas Rao Valluri
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| | - Amit Srivastava
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA; Orbital Therapeutics, Cambridge, MA, USA
| | - Viviane Wal Julião
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| | - Olga Chameh Melone
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| | - Kristen E Allen
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| | - Moe H Kyaw
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| | - Julia Spinardi
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| | | | - John M McLaughlin
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| |
Collapse
|
374
|
Gim H, Lee S, Seo H, Park Y, Chun BC. Effects of Severe Acute Respiratory Syndrome Coronavirus Vaccination on Reinfection: A Community-Based Retrospective Cohort Study. Vaccines (Basel) 2023; 11:1408. [PMID: 37766086 PMCID: PMC10535171 DOI: 10.3390/vaccines11091408] [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: 07/07/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is a disease that is characterized by frequent reinfection. However, the factors influencing reinfection remain poorly elucidated, particularly regarding the effect of COVID-19 vaccination on preventing reinfection and its effects on symptomatology and the interval until reinfection. METHODS This retrospective cohort study examined patients with severe acute respiratory syndrome coronavirus reinfection between January 2020 and February 2022. This study included patients aged >17 years who were reinfected at least 90 days between two infections with severe acute respiratory syndrome coronavirus. The main outcome measure was a reduction in symptoms during reinfection, and reinfection interval. RESULTS Overall, 712 patients (average age: 40.52 ± 16.41 years; 312 males) were included. The reduction rate of symptoms at reinfection than that at first infection was significantly higher in the vaccinated group than in the unvaccinated group (p < 0.001). The average reinfection interval was 265.81 days. The interval between the first and second infection was 63.47 days longer in the vaccinated group than in the unvaccinated group. The interval was also 57.23 days, significantly longer in the asymptomatic group than in the symptomatic group (p < 0.001). CONCLUSIONS Besides its role in preventing severe acute respiratory syndrome coronavirus infection, vaccination reduces the rate of symptomatic reinfection and increases the reinfection interval; thus, it is necessary to be vaccinated even after a previous infection. The findings may inform the decision to avail COVID-19 vaccination.
Collapse
Affiliation(s)
- Hyerin Gim
- Infectious Disease Research Center, Citizens’ Health Bureau, Seoul Metropolitan Government, Seoul 04524, Republic of Korea; (H.G.); (S.L.); (H.S.)
- Department of Epidemiology & Health Informatics, Graduate School of Public Health, Korea University, Seoul 02841, Republic of Korea
| | - Seul Lee
- Infectious Disease Research Center, Citizens’ Health Bureau, Seoul Metropolitan Government, Seoul 04524, Republic of Korea; (H.G.); (S.L.); (H.S.)
| | - Haesook Seo
- Infectious Disease Research Center, Citizens’ Health Bureau, Seoul Metropolitan Government, Seoul 04524, Republic of Korea; (H.G.); (S.L.); (H.S.)
| | - Yumi Park
- Citizens’ Health Bureau, Seoul Metropolitan Government, Seoul 04524, Republic of Korea;
| | - Byung Chul Chun
- Department of Epidemiology & Health Informatics, Graduate School of Public Health, Korea University, Seoul 02841, Republic of Korea
- Department of Preventive Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
375
|
García-Bañuelos J, Oceguera-Contreras E, Sandoval-Rodríguez A, Bastidas-Ramírez BE, Lucano-Landeros S, Gordillo-Bastidas D, Gómez-Meda BC, Santos A, Cerda-Reyes E, Armendariz-Borunda J. AdhMMP8 Vector Administration in Muscle: An Alternate Strategy to Regress Hepatic Fibrosis. Cells 2023; 12:2127. [PMID: 37681859 PMCID: PMC10486800 DOI: 10.3390/cells12172127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/10/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023] Open
Abstract
The development of several vaccines against the SARS-CoV2 virus and their application in millions of people have shown efficacy and safety in the transfer of genes to muscle turning this tissue into a protein-producing factory. Established advanced liver fibrosis, is characterized by replacement of hepatic parenchyma by tissue scar, mostly collagen type I, with increased profibrogenic and proinflammatory molecules gene expression. Matrix metalloproteinase 8 (MMP-8) is an interstitial collagen-degrading proenzyme acting preferentially on collagen type I when activated. This study was carried out to elucidate the effect of an intramuscularly delivered adenoviral vector containing proMMP-8 gene cDNA (AdhMMP8) in male Wistar rats with experimental advanced liver fibrosis induced by thioacetamide. Therapeutic effects were monitored after 1, 2, or 3 weeks of a single dose (3 × 1011 vp/kg) of AdhMMP8. Circulating and liver concentration of MMP-8 protein remained constant; hepatic fibrosis decreased up to 48%; proinflammatory and profibrogenic genes expression diminished: TNF-α 2.28-fold, IL-1 1.95-fold, Col 1A1 4-fold, TGF-β1 3-fold and CTGF 2-fold; and antifibrogenic genes expression raised, MMP-9 2.8-fold and MMP-1 10-fold. Our data proposes that the administration of AdhMMP8 in muscle is safe and effective in achieving liver fibrosis regression at a comparable extent as when the adenoviral vector is delivered systemically to reach the liver, using a minimally invasive procedure.
Collapse
Affiliation(s)
- Jesús García-Bañuelos
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Edén Oceguera-Contreras
- Laboratorio de Sistemas Biológicos, Centro Universitario de los Valles, Universidad de Guadalajara, Carretera Guadalajara-Ameca km. 45.5, Ameca 46600, Jalisco, Mexico
| | - Ana Sandoval-Rodríguez
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Blanca Estela Bastidas-Ramírez
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Silvia Lucano-Landeros
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Daniela Gordillo-Bastidas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Nuevo Leon, Mexico
| | - Belinda C. Gómez-Meda
- Instituto de Genética Humana “Dr. Enrique Corona Rivera”, Department of Molecular Biology and Genomics, Health Sciences University Center, Guadalajara 44340, Jalisco, Mexico
| | - Arturo Santos
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Nuevo Leon, Mexico
| | | | - Juan Armendariz-Borunda
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Nuevo Leon, Mexico
| |
Collapse
|
376
|
Akahata W, Sekida T, Nogimori T, Ode H, Tamura T, Kono K, Kazami Y, Washizaki A, Masuta Y, Suzuki R, Matsuda K, Komori M, Morey AL, Ishimoto K, Nakata M, Hasunuma T, Fukuhara T, Iwatani Y, Yamamoto T, Smith JF, Sato N. Safety and immunogenicity of SARS-CoV-2 self-amplifying RNA vaccine expressing an anchored RBD: A randomized, observer-blind phase 1 study. Cell Rep Med 2023; 4:101134. [PMID: 37586325 PMCID: PMC10439244 DOI: 10.1016/j.xcrm.2023.101134] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/16/2023] [Accepted: 07/07/2023] [Indexed: 08/18/2023]
Abstract
VLPCOV-01 is a lipid nanoparticle-encapsulated self-amplifying RNA (saRNA) vaccine that expresses a membrane-anchored receptor-binding domain (RBD) derived from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. A phase 1 study of VLPCOV-01 is conducted (jRCT2051210164). Participants who completed two doses of the BNT162b2 mRNA vaccine previously are randomized to receive one intramuscular vaccination of 0.3, 1.0, or 3.0 μg VLPCOV-01, 30 μg BNT162b2, or placebo. No serious adverse events have been reported. VLPCOV-01 induces robust immunoglobulin G (IgG) titers against the RBD protein that are maintained up to 26 weeks in non-elderly participants, with geometric means ranging from 5,037 (95% confidence interval [CI] 1,272-19,940) at 0.3 μg to 12,873 (95% CI 937-17,686) at 3 μg compared with 3,166 (95% CI 1,619-6,191) with 30 μg BNT162b2. Neutralizing antibody titers against all variants of SARS-CoV-2 tested are induced. VLPCOV-01 is immunogenic following low-dose administration. These findings support the potential for saRNA as a vaccine platform.
Collapse
Affiliation(s)
- Wataru Akahata
- VLP Therapeutics Japan, Inc., Marunouchi, Minato-ku, Tokyo 105-0003, Japan.
| | - Takashi Sekida
- VLP Therapeutics Japan, Inc., Marunouchi, Minato-ku, Tokyo 105-0003, Japan
| | - Takuto Nogimori
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Hirotaka Ode
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi 460-0001, Japan
| | - Tomokazu Tamura
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kaoru Kono
- VLP Therapeutics Japan, Inc., Marunouchi, Minato-ku, Tokyo 105-0003, Japan
| | - Yoko Kazami
- VLP Therapeutics Japan, Inc., Marunouchi, Minato-ku, Tokyo 105-0003, Japan
| | - Ayaka Washizaki
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Yuji Masuta
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Rigel Suzuki
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | | | - Mai Komori
- VLP Therapeutics, Inc., Gaithersburg, MD 20878, USA
| | | | | | - Misako Nakata
- VLP Therapeutics Japan, Inc., Marunouchi, Minato-ku, Tokyo 105-0003, Japan
| | - Tomoko Hasunuma
- Department of Research, Kitasato University, Kitasato Institute Hospital, Minato-ku, Tokyo 108-0072, Japan
| | - Takasuke Fukuhara
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan; Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yasumasa Iwatani
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi 460-0001, Japan; Division of Basic Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Takuya Yamamoto
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | | | - Nobuaki Sato
- VLP Therapeutics Japan, Inc., Marunouchi, Minato-ku, Tokyo 105-0003, Japan
| |
Collapse
|
377
|
Qureshi A, Connolly JB. Bioinformatic and literature assessment of toxicity and allergenicity of a CRISPR-Cas9 engineered gene drive to control Anopheles gambiae the mosquito vector of human malaria. Malar J 2023; 22:234. [PMID: 37580703 PMCID: PMC10426224 DOI: 10.1186/s12936-023-04665-5] [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] [Received: 12/02/2022] [Accepted: 08/07/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Population suppression gene drive is currently being evaluated, including via environmental risk assessment (ERA), for malaria vector control. One such gene drive involves the dsxFCRISPRh transgene encoding (i) hCas9 endonuclease, (ii) T1 guide RNA (gRNA) targeting the doublesex locus, and (iii) DsRed fluorescent marker protein, in genetically-modified mosquitoes (GMMs). Problem formulation, the first stage of ERA, for environmental releases of dsxFCRISPRh previously identified nine potential harms to the environment or health that could occur, should expressed products of the transgene cause allergenicity or toxicity. METHODS Amino acid sequences of hCas9 and DsRed were interrogated against those of toxins or allergens from NCBI, UniProt, COMPARE and AllergenOnline bioinformatic databases and the gRNA was compared with microRNAs from the miRBase database for potential impacts on gene expression associated with toxicity or allergenicity. PubMed was also searched for any evidence of toxicity or allergenicity of Cas9 or DsRed, or of the donor organisms from which these products were originally derived. RESULTS While Cas9 nuclease activity can be toxic to some cell types in vitro and hCas9 was found to share homology with the prokaryotic toxin VapC, there was no evidence from previous studies of a risk of toxicity to humans and other animals from hCas9. Although hCas9 did contain an 8-mer epitope found in the latex allergen Hev b 9, the full amino acid sequence of hCas9 was not homologous to any known allergens. Combined with a lack of evidence in the literature of Cas9 allergenicity, this indicated negligible risk to humans of allergenicity from hCas9. No matches were found between the gRNA and microRNAs from either Anopheles or humans. Moreover, potential exposure to dsxFCRISPRh transgenic proteins from environmental releases was assessed as negligible. CONCLUSIONS Bioinformatic and literature assessments found no convincing evidence to suggest that transgenic products expressed from dsxFCRISPRh were allergens or toxins, indicating that environmental releases of this population suppression gene drive for malaria vector control should not result in any increased allergenicity or toxicity in humans or animals. These results should also inform evaluations of other GMMs being developed for vector control and in vivo clinical applications of CRISPR-Cas9.
Collapse
Affiliation(s)
- Alima Qureshi
- Department of Life Sciences, Imperial College London, Silwood Park, Sunninghill, Ascot, UK
| | - John B Connolly
- Department of Life Sciences, Imperial College London, Silwood Park, Sunninghill, Ascot, UK.
| |
Collapse
|
378
|
Murphy EA, Guzman-Cardozo C, Sukhu AC, Parks DJ, Prabhu M, Mohammed I, Jurkiewicz M, Ketas TJ, Singh S, Canis M, Bednarski E, Hollingsworth A, Thompson EM, Eng D, Bieniasz PD, Riley LE, Hatziioannou T, Yang YJ. SARS-CoV-2 vaccination, booster, and infection in pregnant population enhances passive immunity in neonates. Nat Commun 2023; 14:4598. [PMID: 37563124 PMCID: PMC10415289 DOI: 10.1038/s41467-023-39989-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/06/2023] [Indexed: 08/12/2023] Open
Abstract
The effects of heterogeneous infection, vaccination and boosting histories prior to and during pregnancy have not been extensively studied and are likely important for protection of neonates. We measure levels of spike binding antibodies in 4600 patients and their neonates with different vaccination statuses, with and without history of SARS-CoV-2 infection. We investigate neutralizing antibody activity against different SARS-CoV-2 variant pseudotypes in a subset of 259 patients and determined correlation between IgG levels and variant neutralizing activity. We further study the ability of maternal antibody and neutralizing measurements to predict neutralizing antibody activity in the umbilical cord blood of neonates. In this work, we show SARS-CoV-2 vaccination and boosting, especially in the setting of previous infection, leads to significant increases in antibody levels and neutralizing activity even against the recent omicron BA.1 and BA.5 variants in both pregnant patients and their neonates.
Collapse
Affiliation(s)
- Elisabeth A Murphy
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, US
| | | | - Ashley C Sukhu
- Department of Pathology and Laboratory Medicine, New York Presbyterian/Weill Cornell Medical Center, New York, NY, US
| | - Debby J Parks
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, US
| | - Malavika Prabhu
- Department of Obstetrics & Gynecology, Weill Cornell Medicine, New York, NY, US
| | - Iman Mohammed
- Department of Obstetrics & Gynecology, Weill Cornell Medicine, New York, NY, US
| | - Magdalena Jurkiewicz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, US
| | - Thomas J Ketas
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, US
| | | | - Marie Canis
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, US
| | - Eva Bednarski
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, US
| | | | | | - Dorothy Eng
- Department of Pathology and Laboratory Medicine, New York Presbyterian/Weill Cornell Medical Center, New York, NY, US
| | - Paul D Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, US
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, US
| | - Laura E Riley
- Department of Obstetrics & Gynecology, Weill Cornell Medicine, New York, NY, US
| | | | - Yawei J Yang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, US.
- Department of Pathology and Laboratory Medicine, New York Presbyterian/Weill Cornell Medical Center, New York, NY, US.
| |
Collapse
|
379
|
Wiśniewski OW, Czyżniewski B, Żukiewicz-Sobczak W, Gibas-Dorna M. Nutritional Behavior in European Countries during COVID-19 Pandemic-A Review. Nutrients 2023; 15:3451. [PMID: 37571387 PMCID: PMC10420667 DOI: 10.3390/nu15153451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/23/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
COVID-19 is highly linked with hyperinflammation and dysfunction of the immune cells. Studies have shown that adequate nutrition, a modifiable factor affecting immunity and limiting systemic inflammation, may play an adjunct role in combating the negative consequences of SARS-CoV-2 infection. Due to the global lockdown conditions, the COVID-19 pandemic has contributed, among others, to restrictions on fresh food availability and changes in lifestyle and eating behaviors. The aim of this paper was to review the data regarding eating habits in European countries within the general population of adults and some specific subpopulations, including obese, diabetic, and psychiatric patients, during the COVID-19 pandemic. The PubMed database and the official websites of medical organizations and associations were searched for the phrases "COVID" and "eating habits". Papers regarding the pediatric population, non-European countries, presenting aggregated data from different countries worldwide, and reviews were excluded. During the COVID-19 pandemic, unhealthy lifestyles and eating behaviors were commonly reported. These included increased snacking, intake of caloric foods, such as sweets, pastries, and beverages, and a decline in physical activity. Data suggest that poor eating habits that create a positive energy balance have persisted over time as an additional post-COVID negative consequence.
Collapse
Affiliation(s)
- Oskar Wojciech Wiśniewski
- Department of Cardiology-Intensive Therapy and Internal Medicine, Poznan University of Medical Sciences, 49 Przybyszewskiego Street, 60-355 Poznan, Poland
- Department of Nutrition and Food, Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland;
| | - Bartłomiej Czyżniewski
- Faculty of Medicine, Collegium Medicum, University of Zielona Gora, 28 Zyty Street, 65-046 Zielona Gora, Poland;
| | - Wioletta Żukiewicz-Sobczak
- Department of Nutrition and Food, Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland;
| | - Magdalena Gibas-Dorna
- Collegium Medicum, Institute of Health Sciences, University of Zielona Gora, 28 Zyty Street, 65-046 Zielona Gora, Poland
| |
Collapse
|
380
|
Ivanko I, Ćelap I, Margetić S, Marijančević D, Josipović J, Gaćina P. Changes in haemostasis and inflammatory markers after mRNA BNT162b2 and vector Ad26.CoV2.S SARS-CoV-2 vaccination. Thromb Res 2023; 228:137-144. [PMID: 37329722 PMCID: PMC10264328 DOI: 10.1016/j.thromres.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/19/2023]
Abstract
INTRODUCTION Reported thromboembolic events after SARS-CoV-2 vaccinations are still raising concerns, predominantly in non-scientific population. The aim of our study was to investigate the differences between haemostasis and inflammatory markers in the subjects vaccinated with mRNA BNT162b2 and vector Ad26.CoV2.S vaccine. MATERIALS AND METHODS The study included 87 subjects vaccinated with mRNA BNT162b2 and 84 with Ad26.CoV2.S vaccine. All the laboratory parameters (TAT, F 1 + 2, IL-6, CRP, big endothelin-1, platelets, fibrinogen, D-dimers, VWF activity) were investigated for the mRNA vaccine at five (before the first dose, 7 and 14 days after the first and second vaccine dose), and three time points (before the first dose, 7 and 14 days after) for the vector vaccine, respectively. All the markers were measured by well-established laboratory methods. RESULTS Our results have shown statistically higher CRP levels in the vector group 7 days after vaccination (P = 0.014). Furthermore, study has revealed statistically significant rise in D-dimers (P = 0.004) between tested time points in both vaccine groups but without clinical repercussions. CONCLUSION Although statistically significant changes in haemostasis markers have been obtained, they remained clinically irrelevant. Thus, our study implicates that there is no plausible scientific evidence of a significant disruption in the coagulation and inflammatory processes after vaccination with BNT162b2 mRNA and Ad26.CoV2.S vector SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- I Ivanko
- Department of Haematology, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia.
| | - I Ćelap
- Department of Clinical Chemistry, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia; Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - S Margetić
- Department of Clinical Chemistry, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia; School of Medicine, Catholic University of Croatia, Zagreb, Croatia
| | - D Marijančević
- Department of Clinical Chemistry, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia; School of Medicine, Catholic University of Croatia, Zagreb, Croatia
| | - J Josipović
- School of Medicine, Catholic University of Croatia, Zagreb, Croatia; Department of Nephrology, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - P Gaćina
- Department of Haematology, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia; School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
381
|
Kim CY, McNeill EN, Young C, King F, Clague M, Caldwell M, Boruah A, Zucker J, Thakur KT. Observational Study of Patients Hospitalized With Neurologic Events After SARS-CoV-2 Vaccination, December 2020-June 2021. Neurol Clin Pract 2023; 13:e200166. [PMID: 37251368 PMCID: PMC10212233 DOI: 10.1212/cpj.0000000000200166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/20/2023] [Indexed: 05/31/2023]
Abstract
Background and Objectives The global spread of the COVID-19 pandemic accelerated the vaccine development time line, regulatory approval, and widespread implementation in the population underscoring the importance of postauthorization/postlicensure vaccine safety surveillance. To monitor for vaccine-related adverse events, we prospectively identified patients hospitalized for prespecified neurologic conditions who received mRNA or adenovirus COVID-19 vaccines and assessed cases for potential risk factors and alternative etiologies of the adverse event. Methods We identified prespecified neurologic conditions in hospitalized individuals within 6 weeks of receipt of a dose of any COVID-19 vaccination between December 11, 2020, and June 22, 2021 (Columbia University Irving Medical Center/New York Presbyterian Hospital, New York City, New York). Clinical data from electronic medical records in these vaccinated patients were reviewed for assessment of contributing risk factors and etiologies for these neurologic conditions by use of a published algorithm. Results Among 3,830 individuals screened for COVID-19 vaccination status and neurologic conditions, 138 (3.6%) cases were included in this study (126 after mRNA and 6 after Janssen vaccines). The 4 most prevalent neurologic syndromes included ischemic stroke (52, 37.7%), encephalopathy (45, 32.6%), seizure (22, 15.9%), and intracranial hemorrhage (ICH) (13, 9.4%). All 138 cases (100%) had 1 or more risk factors and/or evidence for established causes. Metabolic derangement was the most common etiology for seizures (24, 53.3%) and encephalopathy (5, 22.7%) while hypertension was the most significant risk factor in ischemic stroke (45, 86.5%) and ICH cases (4, 30.8%). Discussion All cases in this study were determined to have at least 1 risk factor and/or known etiology accounting for their neurologic syndromes. Our comprehensive clinical review of these cases supports the safety of mRNA COVID-19 vaccines.
Collapse
Affiliation(s)
- Carla Y Kim
- Departments of Neurology (CYK, ENM, CY, FK, M. Clague, M. Caldwell, AB, KTT) and Infectious Disease (JZ), Columbia University Irving Medical Center/New York Presbyterian Hospital
| | - Emily N McNeill
- Departments of Neurology (CYK, ENM, CY, FK, M. Clague, M. Caldwell, AB, KTT) and Infectious Disease (JZ), Columbia University Irving Medical Center/New York Presbyterian Hospital
| | - Casey Young
- Departments of Neurology (CYK, ENM, CY, FK, M. Clague, M. Caldwell, AB, KTT) and Infectious Disease (JZ), Columbia University Irving Medical Center/New York Presbyterian Hospital
| | - Fredrick King
- Departments of Neurology (CYK, ENM, CY, FK, M. Clague, M. Caldwell, AB, KTT) and Infectious Disease (JZ), Columbia University Irving Medical Center/New York Presbyterian Hospital
| | - Madison Clague
- Departments of Neurology (CYK, ENM, CY, FK, M. Clague, M. Caldwell, AB, KTT) and Infectious Disease (JZ), Columbia University Irving Medical Center/New York Presbyterian Hospital
| | - Marissa Caldwell
- Departments of Neurology (CYK, ENM, CY, FK, M. Clague, M. Caldwell, AB, KTT) and Infectious Disease (JZ), Columbia University Irving Medical Center/New York Presbyterian Hospital
| | - Abhilasha Boruah
- Departments of Neurology (CYK, ENM, CY, FK, M. Clague, M. Caldwell, AB, KTT) and Infectious Disease (JZ), Columbia University Irving Medical Center/New York Presbyterian Hospital
| | - Jason Zucker
- Departments of Neurology (CYK, ENM, CY, FK, M. Clague, M. Caldwell, AB, KTT) and Infectious Disease (JZ), Columbia University Irving Medical Center/New York Presbyterian Hospital
| | - Kiran T Thakur
- Departments of Neurology (CYK, ENM, CY, FK, M. Clague, M. Caldwell, AB, KTT) and Infectious Disease (JZ), Columbia University Irving Medical Center/New York Presbyterian Hospital
| |
Collapse
|
382
|
Md Hussin NS, Karuppannan M, Gopalan Y, Tan KM, Gnanasan S. Exploration of non-pharmacological interventions in the management of behavioural and psychological symptoms of dementia. Singapore Med J 2023; 64:497-502. [PMID: 34600449 PMCID: PMC10476919 DOI: 10.11622/smedj.2021125] [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: 01/28/2020] [Accepted: 01/07/2021] [Indexed: 11/18/2022]
Abstract
Introduction Behavioural and psychological symptoms of dementia (BPSD) are considered integral parts of dementia. While pharmacotherapy is reserved for severe symptoms of BPSD, the associated adverse effects can be detrimental. Therefore, non-pharmacological intervention is recommended as the first line of treatment in the management of BPSD. This study aimed to explore the non-pharmacological approaches for the management of BPSD and the strategies and barriers to implementing them in secondary care facilities in Malaysia. Methods A qualitative study design was employed. Data were collected through observations and semi-structured interviews of 12 caregivers and 11 people with dementia (PWD) at seven secondary care facilities. Observations were written in the field notes, and interviews were audio-recorded and transcribed. All data were subjected to thematic analysis. Results Some personalised non-pharmacological interventions, such as physical exercise, music therapy, reminiscence therapy and pet therapy, were conducted in several nursing care centres. Collaborative care from the care providers and family members was found to be an important facilitating factor. The lack of family support led to care providers carrying additional workload beyond their job scope. Other barriers to non-pharmacological interventions were cultural and language differences between the care providers and PWD, inadequate staff numbers and training, and time constraints. Conclusion Although non-pharmacological approaches have been used to some extent in Malaysia, continuous education and training of healthcare providers and the family members of PWD is needed to overcome the challenges to their successful implementation.
Collapse
Affiliation(s)
- Nur Sabiha Md Hussin
- Department of Pharmacy Practice, Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor, Selangor, Malaysia
| | - Mahmathi Karuppannan
- Department of Pharmacy Practice, Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor, Selangor, Malaysia
| | - Yogheswaran Gopalan
- Department of Pharmacy Practice, Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor, Selangor, Malaysia
| | - Kit Mun Tan
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Shubashini Gnanasan
- Department of Pharmacy Practice, Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor, Selangor, Malaysia
| |
Collapse
|
383
|
Zerbo O, Ray GT, Fireman B, Layefsky E, Goddard K, Ross P, Greenberg M, Klein NP. Individual and neighborhood factors associated with being unvaccinated against COVID-19 among pregnant persons. Hum Vaccin Immunother 2023; 19:2256042. [PMID: 37697942 PMCID: PMC10498815 DOI: 10.1080/21645515.2023.2256042] [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] [Received: 06/19/2023] [Revised: 08/04/2023] [Accepted: 09/02/2023] [Indexed: 09/13/2023] Open
Abstract
We investigated whether unvaccinated pregnant persons cluster geographically and determined factors associated with being unvaccinated using spatial and multivariate logistic regression analyses. Pregnant persons with deliveries from December 15, 2020, through September 30, 2022, at Kaiser Permanente Northern California were included. Of the 85,852 pregnant persons in the study, 46.6% were unvaccinated before and during pregnancy. Spatial analysis identified 5 clusters with high prevalence of unvaccinated pregnant persons. Within these clusters, the proportion of unvaccinated varied from 53% to 62% versus 39% outside the clusters. In covariate-adjusted analyses, residence in a cluster increased the odds of being unvaccinated by 1.64 (95% confidence interval (CI): 1.59,1.69). The odds of being unvaccinated increased among those aged 16-24 years (odds ratio [OR] = 2.69, CI: 2.55, 2.83), aged 25-34 years (OR = 1.59, CI: 1.54, 1.64) compared with age ≥ 35 years, black race (OR = 1.45, CI:1.37, 1.54), and subsidized insurance (OR = 1.32, CI: 1.26, 1.38). The odds of being unvaccinated also increased for pregnant persons living in neighborhoods where the proportion of adults with high school education or less was greater than 20%. Geographic clustering of unvaccinated pregnant persons suggests a need for population-specific-interventions to increase vaccine coverage.
Collapse
Affiliation(s)
- Ousseny Zerbo
- Vaccine Study Center, Kaiser Permanente Northern California, Oakland, CA, USA
| | - G. Thomas Ray
- Vaccine Study Center, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Bruce Fireman
- Vaccine Study Center, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Evan Layefsky
- Vaccine Study Center, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Kristin Goddard
- Vaccine Study Center, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Pat Ross
- Vaccine Study Center, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Mara Greenberg
- Obstetrics and Gynecology, Kaiser Permanente Northern California, Oakland, CA, USA
- Regional Perinatal Service Center, Kaiser Permanente Northern California, Santa Clara, CA, USA
| | - Nicola P. Klein
- Vaccine Study Center, Kaiser Permanente Northern California, Oakland, CA, USA
| |
Collapse
|
384
|
Yan L, Liu L. Bibliometric analysis of the highly cited publications in COVID-19 vaccine. Heliyon 2023; 9:e18540. [PMID: 37576281 PMCID: PMC10413004 DOI: 10.1016/j.heliyon.2023.e18540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/14/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has had widespread effects across the globe and continues to affect global public health. This study aims to select and feature highly cited publications on the COVID-19 vaccine. The Web of Science core database was used to extract relevant articles published in recent years. Progress of vaccine studies made in recent two years has mainly focused on the development of different vaccines and the evaluation of their safety and efficacy for population immunity. Clinical trials mainly focusing on the safety and efficacy of diverse vaccines have flourished. Lipid nanoparticle-formulated, nucleoside-modified RNA vaccine and recombinant adenovirus type-5 (26) vectored SARS-CoV-2 vaccines are most commonly studied. Vaccine application-associated challenges mainly include antibody resistance of new variants and unusual severe complications. The correlation between booster immunizations and reinfection is still in the explored state. Currently, antibody resistance of emerging variants is the main vaccine application-associated challenge and the primary reason for vaccine hesitancy. Effective strategies for reinfection prevention are also urgently needed.
Collapse
Affiliation(s)
- Li Yan
- Cadet Company One, Graduate School of Chinese PLA General Hospital, Beijing, 100853, China
| | - Lihua Liu
- Institution of Hospital Management, Department of Medical Innovation and Research, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
385
|
Mohamed S, Chan CK, Tsang CW, Szeto SK, Fong AH, Chan JC, Wong CY. Case Report: Retinal Vasculitis in Two Adolescents After COVID-19 Vaccination. Ocul Immunol Inflamm 2023; 31:1245-1249. [PMID: 36228169 DOI: 10.1080/09273948.2022.2129694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/26/2022] [Accepted: 09/20/2022] [Indexed: 10/17/2022]
Abstract
PURPOSE To report two cases of bilateral retinal vasculitis in adolescents following COVID-19 vaccination. STUDY DESIGN Case report. RESULTS We report the first two cases of retinal vasculitis in adolescents following COVID-19 vaccinations. Both patients received recent second-dose COVID-19 vaccinations (7 weeks and 4 weeks respectively), and presented with bilateral retinal vasculitis and vitritis. Investigations did not reveal other causes of retinal vasculitis. Both patients' retinal vasculitis settled with a short course of oral prednisolone. CONCLUSION Although rare, the temporal association between vaccination, bilateral eye involvement, and the absence of alternative infective or inflammatory causes, makes this a plausible etiology. mRNA vaccinations may cause an autoimmune reaction via host antigenic mimicry, and systemic vasculitis has previously been described. We believe that a short interval between COVID-19 vaccination doses might be a risk factor for the development of retinal vasculitis in adolescents, and clinicians should be aware to elicit vaccination history.
Collapse
Affiliation(s)
- Shaheeda Mohamed
- Hong Kong Eye Hospital, Kowloon, Hong Kong
- Department of Ophthalmology & Visual Sciences, Chinese University of Hong Kong, Hong Kong, Shatin, China
| | - Carmen Km Chan
- Hong Kong Eye Hospital, Kowloon, Hong Kong
- Department of Ophthalmology & Visual Sciences, Chinese University of Hong Kong, Hong Kong, Shatin, China
| | - Chi Wai Tsang
- Hong Kong Eye Hospital, Kowloon, Hong Kong
- Department of Ophthalmology & Visual Sciences, Chinese University of Hong Kong, Hong Kong, Shatin, China
| | - Simon Kh Szeto
- Hong Kong Eye Hospital, Kowloon, Hong Kong
- Department of Ophthalmology & Visual Sciences, Chinese University of Hong Kong, Hong Kong, Shatin, China
| | - Angie Hc Fong
- Hong Kong Eye Hospital, Kowloon, Hong Kong
- Department of Ophthalmology & Visual Sciences, Chinese University of Hong Kong, Hong Kong, Shatin, China
| | - Jason Ck Chan
- Hong Kong Eye Hospital, Kowloon, Hong Kong
- Department of Ophthalmology & Visual Sciences, Chinese University of Hong Kong, Hong Kong, Shatin, China
| | - Cherie Yk Wong
- Hong Kong Eye Hospital, Kowloon, Hong Kong
- Department of Ophthalmology & Visual Sciences, Chinese University of Hong Kong, Hong Kong, Shatin, China
| |
Collapse
|
386
|
Qiao Y, Wotring JW, Zheng Y, Zhang CJ, Zhang Y, Jiang X, Pretto CD, Eyunni S, Parolia A, He T, Cheng C, Cao X, Wang R, Su F, Ellison SJ, Wang Y, Qin J, Yan H, Zhou Q, Ma L, Sexton JZ, Chinnaiyan AM. Proxalutamide reduces SARS-CoV-2 infection and associated inflammatory response. Proc Natl Acad Sci U S A 2023; 120:e2221809120. [PMID: 37459541 PMCID: PMC10372636 DOI: 10.1073/pnas.2221809120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/12/2023] [Indexed: 07/20/2023] Open
Abstract
Early in the COVID-19 pandemic, data suggested that males had a higher risk of developing severe disease and that androgen deprivation therapy might be associated with protection. Combined with the fact that TMPRSS2 (transmembrane serine protease 2), a host entry factor for the SARS-CoV-2 virus, was a well-known androgen-regulated gene, this led to an upsurge of research investigating androgen receptor (AR)-targeting drugs. Proxalutamide, an AR antagonist, was shown in initial clinical studies to benefit COVID-19 patients; however, further validation is needed as one study was retracted. Due to continued interest in proxalutamide, which is in phase 3 trials, we examined its ability to impact SARS-CoV-2 infection and downstream inflammatory responses. Proxalutamide exerted similar effects as enzalutamide, an AR antagonist prescribed for advanced prostate cancer, in decreasing AR signaling and expression of TMPRSS2 and angiotensin-converting enzyme 2 (ACE2), the SARS-CoV-2 receptor. However, proxalutamide led to degradation of AR protein, which was not observed with enzalutamide. Proxalutamide inhibited SARS-CoV-2 infection with an IC50 value of 97 nM, compared to 281 nM for enzalutamide. Importantly, proxalutamide inhibited infection by multiple SARS-CoV-2 variants and synergized with remdesivir. Proxalutamide protected against cell death in response to tumor necrosis factor alpha and interferon gamma, and overall survival of mice was increased with proxalutamide treatment prior to cytokine exposure. Mechanistically, we found that proxalutamide increased levels of NRF2, an essential transcription factor that mediates antioxidant responses, and decreased lung inflammation. These data provide compelling evidence that proxalutamide can prevent SARS-CoV-2 infection and cytokine-induced lung damage, suggesting that promising clinical data may emerge from ongoing phase 3 trials.
Collapse
Affiliation(s)
- Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI48109
| | - Jesse W. Wotring
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI48109
| | - Yang Zheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Charles J. Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI48109
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Xia Jiang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Carla D. Pretto
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI48109
| | - Sanjana Eyunni
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Abhijit Parolia
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Tongchen He
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Caleb Cheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Rui Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Fengyun Su
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Stephanie J. Ellison
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Yini Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
| | - Honghua Yan
- Kintor Pharmaceutical Limited, Suzhou Industrial Park, Suzhuo215123, China
| | - Qianxiang Zhou
- Kintor Pharmaceutical Limited, Suzhou Industrial Park, Suzhuo215123, China
| | - Liandong Ma
- Kintor Pharmaceutical Limited, Suzhou Industrial Park, Suzhuo215123, China
| | - Jonathan Z. Sexton
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI48109
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI48109
- Center for Drug Repurposing, University of Michigan, Ann Arbor, MI48109
- Michigan Institute for Clinical and Health Research, University of Michigan, Ann Arbor, MI48109
- Department of Pharmacology, University of Michigan, Ann Arbor, MI48109
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI48109
- HHMI, University of Michigan, Ann Arbor, MI48109
- Department of Urology, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
387
|
Turley CB, Tables L, Fuller T, Sanders LJ, Scott H, Moodley A, Woodward Davis A, Leav B, Miller J, Schoemaker K, Vandebosch A, Sadoff J, Woo W, Cho I, Dunkle LM, Li S, van der Laan L, Gilbert PB, Follmann D, Jaynes H, Kublin JG, Baden LR, Goepfert P, Kotloff K, Gay CL, Falsey AR, El Sahly HM, Sobieszczyk ME, Huang Y, Neuzil KM, Corey L, Grinsztejn B, Gray G, Rouphael N, Luedtke A. Modifiers of COVID-19 vaccine efficacy: Results from four COVID-19 prevention network efficacy trials. Vaccine 2023; 41:4899-4906. [PMID: 37385888 PMCID: PMC10288314 DOI: 10.1016/j.vaccine.2023.06.066] [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] [Received: 04/16/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023]
Abstract
Questions remain regarding the effect of baseline host and exposure factors on vaccine efficacy (VE) across pathogens and vaccine platforms. We report placebo-controlled data from four Phase 3 COVID-19 trials during the early period of the pandemic. This was a cross-protocol analysis of four randomized, placebo-controlled efficacy trials (Moderna/mRNA1273, AstraZeneca/AZD1222, Janssen/Ad26.COV2.S, and Novavax/NVX-CoV2373) using a harmonized design. Trials were conducted in the United States and international sites in adults ≥ 18 years of age. VE was assessed for symptomatic and severe COVID-19. We analyzed 114,480 participants from both placebo and vaccine arms, enrolled July 2020 to February 2021, with follow up through July 2021. VE against symptomatic COVID-19 showed little heterogeneity across baseline socio-demographic, clinical or exposure characteristics, in either univariate or multivariate analysis, regardless of vaccine platform. Similarly, VE against severe COVID-19 in the single trial (Janssen) with sufficient endpoints for analysis showed little evidence of heterogeneity. COVID-19 VE is not influenced by baseline host or exposure characteristics across efficacy trials of different vaccine platforms and countries when well matched to circulating virus strains. This supports use of these vaccines, regardless of platform type, as effective tools in the near term for reducing symptomatic and severe COVID-19, particularly for older individuals and those with common co-morbidities during major variant shifts. Clinical trial registration numbers: NCT04470427, NCT04516746, NCT04505722, and NCT04611802.
Collapse
Affiliation(s)
- Christine B Turley
- Atrium Health Wake Forest School of Medicine, Charlotte, NC, United States
| | - LaKesha Tables
- Morehouse School of Medicine, Atlanta, GA, United States
| | - Trevon Fuller
- Infectious Diseases Department, Hospital Federal dos Servidores do Estado, Rio de Janeiro, RJ, Brazil
| | - Lisa J Sanders
- University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Hyman Scott
- San Francisco Department of Public Health, San Francisco, CA, United States
| | - Amaran Moodley
- Division of Infectious Diseases, University of California San Diego and Rady Children's Hospital, San Diego, CA, United States
| | - Amanda Woodward Davis
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Brett Leav
- Moderna Inc., Cambridge, MA, United States
| | | | - Kathryn Schoemaker
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - An Vandebosch
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | - Wayne Woo
- Novavax, Gaithersburg, MD, United States
| | - Iksung Cho
- Novavax, Gaithersburg, MD, United States
| | | | - Sijia Li
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Lars van der Laan
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States
| | - Holly Jaynes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | | | - Paul Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Karen Kotloff
- Division of Infectious Disease and Tropical Pediatrics, Department of Pediatrics, and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Cynthia L Gay
- Department of Medicine, Division of Infectious Diseases, UNC HIV Cure Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Ann R Falsey
- Department of Medicine, Infectious Disease Division, University of Rochester, Rochester, NY, United States
| | - Hana M El Sahly
- Department of Molecular Virology and Microbiology and Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Magdalena E Sobieszczyk
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Kathleen M Neuzil
- Division of Infectious Disease and Tropical Pediatrics, Department of Pediatrics, and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Beatriz Grinsztejn
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Glenda Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; South African Medical Research Council, Cape Town, South Africa
| | | | - Alex Luedtke
- Department of Biostatistics, University of Washington, Seattle, WA, United States.
| |
Collapse
|
388
|
Moll MEC, Martínez AMS, Cisneros BT, Onofre JIG, Floriano GN, de León MB. Side Effects of COVID-19 Vaccines in Pregnant and Lactating Mexican Women and Breastfed Infants: A Survey-Based Study. Vaccines (Basel) 2023; 11:1280. [PMID: 37631848 PMCID: PMC10459654 DOI: 10.3390/vaccines11081280] [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: 05/16/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 08/27/2023] Open
Abstract
COVID-19 vaccines' safety has been extensively studied; however, further analysis is required in pregnant women, nursing mothers, and breastfed infants. Our aim was to compare the extension and severity of self-reported COVID-19 vaccine side effects in pregnant and breastfeeding women, and breastfed infants. In this cross-sectional study, COVID-19-vaccinated subjects were enrolled using an online survey in Mexico. Women were classified by pregnancy and breastfeeding status at the time of vaccination (n = 3167). After the first or only dose, there was a trend toward fewer systemic effects in pregnant women (p = 0.06). BNT162b2 (Pfizer-BioNTech) had a higher frequency of local symptoms in pregnancy. Lactating women experienced fewer local symptoms after the first or single dose (p = 0.04) and the opposite occurred after the second dose (p = 0.001). ChAdOx1 (AstraZeneca) increased the chances of developing both local and systemic symptoms after the first dose but decreased them after the second dose. The severity was similar across groups, although the result of lack of association in pregnancy requires studies with a larger sample size. Irritability was the most reported symptom in breastfed infants. This study contributes to the knowledge about the side effects in pregnant and lactating women, and breastfed babies.
Collapse
Affiliation(s)
- María Elena Camacho Moll
- Department of Molecular Biology, Northeast Biomedical Research Center, Mexican Institute of Social Security, Monterrey 64720, N.L., Mexico;
- Center for Molecular Diagnosis and Personalized Medicine, Health Sciences Division, Universidad de Monterrey, San Pedro Garza García 66238, N.L., Mexico
| | - Ana María Salinas Martínez
- Epidemiologic and Health Services Research Unit, Mexican Institute of Social Security, Monterrey 64360, N.L., Mexico
- School of Public Health and Nutrition, Autonomous University of Nuevo Leon, Monterrey 64460, N.L., Mexico
| | - Benjamín Tovar Cisneros
- School of Biological Sciences, Autonomous University of Nuevo Leon, Monterrey 66455, N.L., Mexico;
| | - Juan Ignacio García Onofre
- Family Medicine Unit No. 64, Mexican Institute of Social Security, Santa Catarina 66358, N.L., Mexico; (J.I.G.O.); (G.N.F.)
| | - Gloria Navarrete Floriano
- Family Medicine Unit No. 64, Mexican Institute of Social Security, Santa Catarina 66358, N.L., Mexico; (J.I.G.O.); (G.N.F.)
| | - Mario Bermúdez de León
- Department of Molecular Biology, Northeast Biomedical Research Center, Mexican Institute of Social Security, Monterrey 64720, N.L., Mexico;
| |
Collapse
|
389
|
Lee CYS, Suzuki JB. COVID-19: Variants, Immunity, and Therapeutics for Non-Hospitalized Patients. Biomedicines 2023; 11:2055. [PMID: 37509694 PMCID: PMC10377623 DOI: 10.3390/biomedicines11072055] [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: 03/14/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The continuing transmission of coronavirus disease 2019 (COVID-19) remains a world-wide 21st-century public health emergency of concern. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused greater than 600 million cases of COVID-19 and over 6 million deaths globally. COVID-19 continues to be a highly transmissible disease despite efforts by public health officials and healthcare providers to manage and control the disease. Variants identified in selected worldwide epicenters add to the complexity of vaccine efficacy, overage, and antibody titer maintenance and bioactivity. The identification of the SARS-CoV-2 variants is described with respect to evading protective efficacy of COVID-19 vaccines and breakthrough infections. Vaccines and other therapeutics have prevented millions of SARS-CoV-2 infections and thousands of deaths in the United States. We explore aspects of the immune response in a condensed discussion to understand B and T cell lymphocyte regulatory mechanisms and antibody effectiveness and senescence. Finally, COVID-19 therapies including Paxlovid, Remdisivir, Molnupiravir and convalescent plasma in non-hospitalized patients are presented with limitations for identification, collection, and distribution to infected patients.
Collapse
Affiliation(s)
- Cameron Y S Lee
- Private Practice in Oral, Maxillofacial and Reconstructive Surgery, Aiea, HI 96701, USA
- Department of Periodontology and Oral Implantology, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19140, USA
| | - Jon B Suzuki
- Department of Periodontology and Oral Implantology, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19140, USA
- Department of Graduate Periodontics, University of Maryland, Baltimore, MD 20742, USA
- Department of Graduate Prosthodontics, University of Washington, Seattle, WA 98195, USA
- Department of Graduate Periodontics, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
- Department of Microbiology and Immunology, School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
390
|
Mueller-Enz M, Woopen C, Katoul Al Rahbani G, Haase R, Dunsche M, Ziemssen T, Akgün K. NVX-CoV2373-induced T- and B-cellular immunity in immunosuppressed people with multiple sclerosis that failed to respond to mRNA and viral vector SARS-CoV-2 vaccines. Front Immunol 2023; 14:1081933. [PMID: 37545513 PMCID: PMC10399811 DOI: 10.3389/fimmu.2023.1081933] [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: 10/27/2022] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
Importance Immunological response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination is important, especially in people with multiple sclerosis (pwMS) on immunosuppressive therapies. Objective This study aims to determine whether adjuvanted protein-based vaccine NVX-CoV2373 is able to induce an immune response to SARS-CoV-2 in pwMS with inadequate responses to prior triple mRNA/viral vector vaccination. Design setting and participants We conducted a single-center, prospective longitudinal cohort study at the MS Center in Dresden, Germany. In total, 65 participants were included in the study in accordance with the following eligibility criteria: age > 18 years, immunomodulatory treatment, and insufficient T-cellular and humoral response to prior vaccination with at least two doses of SARS-CoV-2 mRNA (BNT162b2, mRNA-1273) or viral vector vaccines (AZD1222, Ad26.COV2.S). Interventions Intramuscular vaccination with two doses of NVX-CoV2373 at baseline and 3 weeks of follow-up. Main outcomes and measures The development of SARS-CoV-2-specific antibodies and T-cell responses was evaluated. Results For the final analysis, data from 47 patients on stable treatment with sphingosine-1-phosphate receptor (S1PR) modulators and 17 on ocrelizumab were available. The tolerability of the NVX-CoV2373 vaccination was overall good and comparable to the one reported for the general population. After the second NVX-CoV2373 vaccination, 59% of S1PR-modulated patients developed antispike IgG antibodies above the predefined cutoff of 200 binding antibody units (BAU)/ml (mean, 1,204.37 [95% CI, 693.15, 2,092.65] BAU/ml), whereas no clinically significant T-cell response was found. In the subgroup of the patients on ocrelizumab treatment, 23.5% developed antispike IgG > 200 BAU/ml (mean, 116.3 [95% CI, 47.04, 287.51] BAU/ml) and 53% showed positive spike-specific T-cellular responses (IFN-gamma release to antigen 1: mean, 0.2 [95% CI, 0.11, 0.31] IU/ml; antigen 2: mean, 0.24 [95% CI, 0.14, 0.37]) after the second vaccination. Conclusions Vaccination with two doses of NVX-CoV2373 was able to elicit a SARS-CoV-2-specific immune response in pwMS lacking adequate immune responses to previous mRNA/viral vector vaccination. For patients receiving S1PR modulators, an increase in anti-SARS-CoV-2 IgG antibodies was detected after NVX-CoV2373 vaccination, whereas in ocrelizumab-treated patients, the increase of antiviral T-cell responses was more pronounced. Our data may impact clinical decision-making by influencing the preference for NVX-CoV2373 vaccination in pwMS receiving treatment with S1PR modulation or anti-CD20 treatment.
Collapse
|
391
|
Muangnoicharoen S, Wiangcharoen R, Nanthapisal S, Kamolratakul S, Lawpoolsri S, Jongkaewwattana A, Thitithanyanont A, Luvira V, Chinwangso P, Thanthamnu N, Chantratita N, Lim JK, Anh Wartel T, Excler JL, Ryser MF, Leong C, Mak TK, Pitisuttithum P. Single Ad26.COV2.S booster dose following two doses of BBIBP-CorV vaccine against SARS-CoV-2 infection in adults: Day 28 results of a phase 1/2 open-label trial. Vaccine 2023; 41:4648-4657. [PMID: 37344265 PMCID: PMC10267503 DOI: 10.1016/j.vaccine.2023.06.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/12/2023] [Accepted: 06/12/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND The inactivated COVID-19 whole-virus vaccine BBIBP-CorV has been extensively used worldwide. Heterologous boosting after primary vaccination can induce higher immune responses against SARS-CoV-2 than homologous boosting. The safety and immunogenicity after 28 days of a single Ad26.COV2.S booster dose given at different intervals after 2 doses of BBIBP-CorV are presented. METHODS This open-label phase 1/2 trial was conducted in healthy adults in Thailand who had completed 2-dose primary vaccination with BBIBP-CorV. Participants received a single booster dose of Ad26.COV2.S (5 × 1010 virus particles) 90-240 days (Group A1; n = 360) or 45-75 days (Group A2; n = 66) after the second BBIBP-CorV dose. Safety and immunogenicity were assessed over 28 days. Binding IgG antibodies to the full-length pre-fusion Spike and anti-nucleocapsid proteins of SARS-CoV-2 were measured by enzyme-linked immunosorbent assay. The SARS-CoV-2 pseudovirus neutralization assay and live virus microneutralization assay were used to quantify the neutralizing activity of antibodies against ancestral SARS-CoV-2 (Wuhan-Hu-1) and the delta (B.1.617.2) and omicron (B.1.1.529/BA.1 and BA.2) variants. The cell-mediated immune response was measured using a quantitative interferon (IFN)-γ release assay in whole blood. RESULTS Solicited local and systemic adverse events (AEs) on days 0-7 were mostly mild, as were unsolicited vaccine-related AEs during days 0-28, with no serious AEs. On day 28, anti-Spike binding antibodies increased from baseline by 487- and 146-fold in Groups A1 and A2, and neutralizing antibodies against ancestral SARS-CoV-2 by 55- and 37-fold, respectively. Humoral responses were strongest against ancestral SARS-CoV-2, followed by the delta, then the omicron BA.2 and BA.1 variants. T-cell-produced interferon-γ increased approximately 10-fold in both groups. CONCLUSIONS A single heterologous Ad26.COV2.S booster dose after two BBIBP-CorV doses was well tolerated and induced robust humoral and cell-mediated immune responses measured at day 28 in both interval groups. CLINICAL TRIALS REGISTRATION NCT05109559.
Collapse
Affiliation(s)
- Sant Muangnoicharoen
- Vaccine Trial Centre, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Rakpong Wiangcharoen
- Phaholpolpayuhasena Hospital, 572 Saeng Chuto Road Muang, Kanchanaburi 71000, Thailand
| | - Sira Nanthapisal
- Faculty of Medicine, Thammasat University (Rangsit Campus), Pathum Thani, Thailand
| | - Supitcha Kamolratakul
- Vaccine Trial Centre, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Saranath Lawpoolsri
- Center of Excellence for Biomedical and Public Health Informatics (BIOPHICS), Bangkok, Thailand; Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Anan Jongkaewwattana
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | | | - Viravarn Luvira
- Vaccine Trial Centre, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Pailinrut Chinwangso
- Center of Excellence for Biomedical and Public Health Informatics (BIOPHICS), Bangkok, Thailand
| | - Narumon Thanthamnu
- Vaccine Trial Centre, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | | | | | - T Anh Wartel
- International Vaccine Institute, Seoul, Republic of Korea
| | | | | | - Chloe Leong
- Janssen Asia Pacific Medical Affairs Operations, Sydney, Australia
| | - Tippi K Mak
- Centre of Regulatory Excellence, Duke-NUS Medical School, Singapore; Vaccine and Infectious Disease Organization, University of Saskatchewan, Canada
| | - Punnee Pitisuttithum
- Vaccine Trial Centre, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand.
| |
Collapse
|
392
|
Tavazzi E, Pichiecchio A, Colombo E, Rigoni E, Asteggiano C, Vegezzi E, Masi F, Greco G, Bastianello S, Bergamaschi R. The Potential Role of SARS-CoV-2 Infection and Vaccines in Multiple Sclerosis Onset and Reactivation: A Case Series and Literature Review. Viruses 2023; 15:1569. [PMID: 37515255 PMCID: PMC10385211 DOI: 10.3390/v15071569] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
The recent SARS-CoV-2 pandemic and related vaccines have raised several issues. Among them, the potential role of the viral infection (COVID-19) or anti-SARS-CoV-2 vaccines as causal factors of dysimmune CNS disorders, as well as the safety and efficacy of vaccines in patients affected by such diseases and on immune-active treatments have been analyzed. The aim is to better understand the relationship between SARS-CoV-2 infection/vaccines with dysimmune CNS diseases by describing 12 cases of multiple sclerosis/myelitis onset or reactivation after exposure to SARS-CoV-2 infection/vaccines and reviewing all published case reports or case series in which MS onset or reactivation was temporally associated with either COVID-19 (8 case reports, 3 case series) or anti-SARS-CoV-2 vaccines (13 case reports, 6 case series). All the cases share a temporal association between viral/vaccine exposure and symptoms onset. This finding, together with direct or immune-based mechanisms described both during COVID-19 and MS, claims in favor of a role for SARS-CoV-2 infection/vaccines in unmasking dysimmune CNS disorders. The most common clinical presentations involve the optic nerve, brainstem and spinal cord. The preferential tropism of the virus together with the presence of some host-related genetic/immune factors might predispose to the involvement of specific CNS districts.
Collapse
Affiliation(s)
| | - Anna Pichiecchio
- IRCCS Mondino Foundation, 27100 Pavia, Italy
- Department of Brain and Behavioural Sciences, University of Pavia, 27100 Pavia, Italy
| | | | | | - Carlo Asteggiano
- IRCCS Mondino Foundation, 27100 Pavia, Italy
- Department of Brain and Behavioural Sciences, University of Pavia, 27100 Pavia, Italy
| | | | - Francesco Masi
- IRCCS Mondino Foundation, 27100 Pavia, Italy
- Department of Brain and Behavioural Sciences, University of Pavia, 27100 Pavia, Italy
| | - Giacomo Greco
- IRCCS Mondino Foundation, 27100 Pavia, Italy
- Department of Brain and Behavioural Sciences, University of Pavia, 27100 Pavia, Italy
| | - Stefano Bastianello
- Department of Brain and Behavioural Sciences, University of Pavia, 27100 Pavia, Italy
| | | |
Collapse
|
393
|
Gilbert-Esparza E, Brady A, Haas S, Wittstruck H, Miller J, Kang Q, Mulcahy ER. Vaccine Hesitancy in College Students. Vaccines (Basel) 2023; 11:1243. [PMID: 37515058 PMCID: PMC10385752 DOI: 10.3390/vaccines11071243] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
(1) Background: Following the COVID-19 pandemic, vaccine hesitancy has become an increasingly important topic and has created significant concerns in public health. It is important to explore vaccine hesitancy among college students as they have been identified to be a high-risk group for COVID-19 transmission. This study aims to investigate COVID-19 vaccine hesitancy in college students on a midsized midwestern university campus. (2) Methods: Data were collected from 311 undergraduate and graduate college students during June and July 2021. Participants completed a survey on COVID-19 vaccine behaviors, perceptions, and opinions. Quantitative and qualitative analysis was performed to identify vaccine hesitancy and influencing factors in the student population. (3) Results: The results of this study demonstrated significant relationships between older and younger undergraduate students (OR > 1, p < 0.05), students who received a yearly influenza vaccine and those that did not (p < 0.05), and students who had a previous COVID-19 infection and those that did not (OR > 1, p < 0.05). We also determined a significant difference between some racial/ethnic groups and vaccine hesitancy status. (4) Conclusions: COVID-19 vaccine hesitancy exists on college campuses, and is influenced by age and student status, influenza vaccination status, previous COVID-19 infection, and race/ethnicity.
Collapse
Affiliation(s)
| | - Amelia Brady
- Master of Public Health Program, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Sierrah Haas
- Master of Public Health Program, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Heather Wittstruck
- Master of Public Health Program, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Jennifer Miller
- Kansas Department of Health and Environment, Topeka, KS 66612, USA
| | - Qing Kang
- Department of Statistics, Kansas State University, Manhattan, KS 66506, USA
| | - Ellyn R Mulcahy
- Master of Public Health Program, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
394
|
Babu TM, Feldstein LR, Saydah S, Acker Z, Boisvert CL, Briggs-Hagen M, Carone M, Casto A, Cox SN, Ehmen B, Englund JA, Fortmann SP, Frivold CJ, Groom H, Han PD, Kuntz JL, Lockwood T, Midgley CM, Mularski RA, Ogilvie T, Reich SL, Schmidt MA, Smith N, Starita L, Stone J, Vandermeer M, Weil AA, Wolf CR, Chu HY, Naleway AL. CASCADIA: a prospective community-based study protocol for assessing SARS-CoV-2 vaccine effectiveness in children and adults using a remote nasal swab collection and web-based survey design. BMJ Open 2023; 13:e071446. [PMID: 37451722 PMCID: PMC10350906 DOI: 10.1136/bmjopen-2022-071446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
INTRODUCTION Although SARS-CoV-2 vaccines were first approved under Emergency Use Authorization by the Food and Drug Administration in late 2020 for adults, authorisation for young children 6 months to <5 years of age did not occur until 2022. These authorisations were based on clinical trials, understanding real-world vaccine effectiveness (VE) in the setting of emerging variants is critical. The primary goal of this study is to evaluate SARS-CoV-2 VE against infection among children aged >6 months and adults aged <50 years. METHODS CASCADIA is a 4-year community-based prospective study of SARS-CoV-2 VE among 3500 adults and paediatric populations aged 6 months to 49 years in Oregon and Washington, USA. At enrolment and regular intervals, participants complete a sociodemographic questionnaire. Individuals provide a blood sample at enrolment and annually thereafter, with optional blood draws every 6 months and after infection and vaccination. Participants complete weekly self-collection of anterior nasal swabs and symptom questionnaires. Swabs are tested for SARS-CoV-2 and other respiratory pathogens by reverse transcription-PCR, with results of selected pathogens returned to participants; nasal swabs with SARS-CoV-2 detected will undergo whole genome sequencing. Participants who test positive for SARS-CoV-2 undergo serial swab collection every 3 days for 21 days. Serum samples are tested for SARS-CoV-2 antibody by binding and neutralisation assays. ANALYSIS The primary outcome is SARS-CoV-2 infection. Cox regression models will be used to estimate the incidence rate ratio associated with SARS-CoV-2 vaccination among the paediatric and adult population, controlling for demographic factors and other potential confounders. ETHICS AND DISSEMINATION All study materials including the protocol, consent forms, data collection instruments, participant communication and recruitment materials, were approved by the Kaiser Permanente Interregional Institutional Review Board, the IRB of record for the study. Results will be disseminated through peer-reviewed publications, presentations, participant newsletters and appropriate general news media.
Collapse
Affiliation(s)
- Tara M Babu
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Leora R Feldstein
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Sharon Saydah
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Zachary Acker
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, Washington, USA
| | | | - Melissa Briggs-Hagen
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Marco Carone
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Amanda Casto
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Sarah N Cox
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Brenna Ehmen
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, Washington, USA
| | - Janet A Englund
- Department of Pediatrics, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Stephen P Fortmann
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, USA
| | - Collrane J Frivold
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Holly Groom
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, USA
| | - Peter D Han
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, Washington, USA
| | - Jennifer L Kuntz
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, USA
| | - Tina Lockwood
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, Washington, USA
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Claire M Midgley
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Richard A Mularski
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, USA
| | - Tara Ogilvie
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Sacha L Reich
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, USA
| | - Mark A Schmidt
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, USA
| | - Ning Smith
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, USA
| | - Lea Starita
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, Washington, USA
- Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Jeremy Stone
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, Washington, USA
| | - Meredith Vandermeer
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, USA
| | - Ana A Weil
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Caitlin R Wolf
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Helen Y Chu
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Allison L Naleway
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, USA
| |
Collapse
|
395
|
Warpechowski J, Leszczyńska P, Juchnicka D, Olichwier A, Szczerbiński Ł, Krętowski AJ. Assessment of the Immune Response in Patients with Insulin Resistance, Obesity, and Diabetes to COVID-19 Vaccination. Vaccines (Basel) 2023; 11:1203. [PMID: 37515018 PMCID: PMC10383449 DOI: 10.3390/vaccines11071203] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
The SARS-CoV-19 pandemic overwhelmed multiple healthcare systems across the world. Patients with underlying medical conditions such as obesity or diabetes were particularly vulnerable, had more severe symptoms, and were more frequently hospitalized. To date, there have been many studies on the severity of SARS-CoV-2 in patients with metabolic disorders, but data on the efficiency of vaccines against COVID-19 are still limited. This paper aims to provide a comprehensive overview of the effectiveness of COVID-19 vaccines in individuals with diabetes, insulin resistance, and obesity. A comparison is made between the immune response after vaccination in patients with and without metabolic comorbidities. Additionally, an attempt is made to highlight the mechanisms of immune stimulation affected by SARS-CoV-2 vaccines and how metabolic comorbidities modulate these mechanisms. The focus is on the most common COVID-19 vaccines, which include mRNA vaccines such as Pfizer-BioNTech and Moderna, as well as viral vector vaccines such as AstraZeneca and Johnson & Johnson. Furthermore, an effort is made to clarify how the functional differences between these vaccines may impact the response in individuals with metabolic disorders, drawing from available experimental data. This review summarizes the current knowledge regarding the post-vaccination response to COVID-19 in the context of metabolic comorbidities such as diabetes, insulin resistance, and obesity.
Collapse
Affiliation(s)
- Jędrzej Warpechowski
- Clinical Research Centre, Medical University of Bialystok, Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
| | - Paula Leszczyńska
- Clinical Research Centre, Medical University of Bialystok, Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
| | - Dominika Juchnicka
- Clinical Research Centre, Medical University of Bialystok, Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
| | - Adam Olichwier
- Clinical Research Centre, Medical University of Bialystok, Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Łukasz Szczerbiński
- Clinical Research Centre, Medical University of Bialystok, Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
- Department of Endocrinology, Diabetology and Internal Diseases, Medical University of Bialystok, Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
- Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
| | - Adam Jacek Krętowski
- Clinical Research Centre, Medical University of Bialystok, Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
- Department of Endocrinology, Diabetology and Internal Diseases, Medical University of Bialystok, Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
| |
Collapse
|
396
|
Mabuka T, Naidoo N, Ncube N, Yiga T, Ross M, Kurehwa K, Nare Nyathi M, Silaji A, Ndemera T, Lemeke T, Taiwo R, Macharia W, Sithole M. The Impact of SARS-CoV-2 Lineages (Variants) and COVID-19 Vaccination on the COVID-19 Epidemic in South Africa: Regression Study. JMIRX MED 2023; 4:e34598. [PMID: 37463043 PMCID: PMC10337479 DOI: 10.2196/34598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 04/12/2022] [Accepted: 02/13/2023] [Indexed: 07/20/2023]
Abstract
Background Emerging SARS-CoV-2 variants have been attributed to the occurrence of secondary, tertiary, quaternary, and quinary COVID-19 epidemic waves threatening vaccine efforts owing to their immune invasiveness. Since the importation of SARS-CoV-2 in South Africa, with the first reported COVID-19 case on March 5, 2020, South Africa has observed 5 consecutive COVID-19 epidemic waves. The evolution of SARS-CoV-2 has played a major role in the resurgence of COVID-19 epidemic waves in South Africa and across the globe. Objective We aimed to conduct descriptive and inferential statistical analysis on South African COVID-19 epidemiological data to investigate the impact of SARS-CoV-2 lineages and COVID-19 vaccinations in South African COVID-19 epidemiology. Methods The general methodology involved the collation and stratification, covariance, regression analysis, normalization, and comparative inferential statistical analysis through null hypothesis testing (paired 2-tailed t tests) of South African COVID-19 epidemiological data. Results The mean daily positive COVID-19 tests in South Africa's first, second, third, fourth, and fifth COVID-19 epidemic wave periods were 11.5% (SD 8.58%), 11.5% (SD 8.45%), 13.3% (SD 9.72%), 13.1% (SD 9.91%), and 14.3% (SD 8.49%), respectively. The COVID-19 transmission rate in the first and second COVID-19 epidemic waves in South Africa was similar, while the COVID-19 transmission rate was higher in the third, fourth, and fifth COVID-19 epidemic waves than in the aforementioned waves. Most COVID-19 hospitalized cases in South Africa were in the general ward (60%-79.1%). Patients with COVID-19 on oxygen were the second-largest admission status (11.2%-16.8%), followed by patients with COVID-19 in the intensive care unit (8.07%-16.7%). Most patients hospitalized owing to COVID-19 in South Africa's first, second, third, and fourth COVID-19 epidemic waves were aged between 40 and 49 years (16.8%-20.4%) and 50 and 59 years (19.8%-25.3%). Patients admitted to the hospital owing to COVID-19 in the age groups of 0 to 19 years were relatively low (1.98%-4.59%). In general, COVID-19 hospital admissions in South Africa for the age groups between 0 and 29 years increased after each consecutive COVID-19 epidemic wave, while for age groups between 30 and 79 years, hospital admissions decreased. Most COVID-19 hospitalization deaths in South Africa in the first, second, third, fourth, and fifth COVID-19 epidemic waves were in the ages of 50 to 59 years (15.8%-24.8%), 60 to 69 years (15.9%-29.5%), and 70 to 79 years (16.6%-20.7%). Conclusions The relaxation of COVID-19 nonpharmaceutical intervention health policies in South Africa and the evolution of SARS-CoV-2 were associated with increased COVID-19 transmission and severity in the South African population. COVID-19 vaccination in South Africa was strongly associated with a decrease in COVID-19 hospitalization and severity in South Africa.
Collapse
Affiliation(s)
- Thabo Mabuka
- The Afrikan Research Initiative Motloung South Africa
| | | | - Nesisa Ncube
- The Afrikan Research Initiative Motloung South Africa
| | - Thabo Yiga
- The Afrikan Research Initiative Motloung South Africa
| | - Michael Ross
- The Afrikan Research Initiative Motloung South Africa
| | | | | | - Andrea Silaji
- The Afrikan Research Initiative Motloung South Africa
| | | | | | - Ridwan Taiwo
- The Afrikan Research Initiative Motloung South Africa
| | | | | |
Collapse
|
397
|
Theodore DA, Branche AR, Zhang L, Graciaa DS, Choudhary M, Hatlen TJ, Osman R, Babu TM, Robinson ST, Gilbert PB, Follmann D, Janes H, Kublin JG, Baden LR, Goepfert P, Gray GE, Grinsztejn B, Kotloff KL, Gay CL, Leav B, Miller J, Hirsch I, Sadoff J, Dunkle LM, Neuzil KM, Corey L, Falsey AR, El Sahly HM, Sobieszczyk ME, Huang Y. Clinical and Demographic Factors Associated With COVID-19, Severe COVID-19, and SARS-CoV-2 Infection in Adults: A Secondary Cross-Protocol Analysis of 4 Randomized Clinical Trials. JAMA Netw Open 2023; 6:e2323349. [PMID: 37440227 PMCID: PMC10346130 DOI: 10.1001/jamanetworkopen.2023.23349] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/15/2023] [Indexed: 07/14/2023] Open
Abstract
Importance Current data identifying COVID-19 risk factors lack standardized outcomes and insufficiently control for confounders. Objective To identify risk factors associated with COVID-19, severe COVID-19, and SARS-CoV-2 infection. Design, Setting, and Participants This secondary cross-protocol analysis included 4 multicenter, international, randomized, blinded, placebo-controlled, COVID-19 vaccine efficacy trials with harmonized protocols established by the COVID-19 Prevention Network. Individual-level data from participants randomized to receive placebo within each trial were combined and analyzed. Enrollment began July 2020 and the last data cutoff was in July 2021. Participants included adults in stable health, at risk for SARS-CoV-2, and assigned to the placebo group within each vaccine trial. Data were analyzed from April 2022 to February 2023. Exposures Comorbid conditions, demographic factors, and SARS-CoV-2 exposure risk at the time of enrollment. Main Outcomes and Measures Coprimary outcomes were COVID-19 and severe COVID-19. Multivariate Cox proportional regression models estimated adjusted hazard ratios (aHRs) and 95% CIs for baseline covariates, accounting for trial, region, and calendar time. Secondary outcomes included severe COVID-19 among people with COVID-19, subclinical SARS-CoV-2 infection, and SARS-CoV-2 infection. Results A total of 57 692 participants (median [range] age, 51 [18-95] years; 11 720 participants [20.3%] aged ≥65 years; 31 058 participants [53.8%] assigned male at birth) were included. The analysis population included 3270 American Indian or Alaska Native participants (5.7%), 7849 Black or African American participants (13.6%), 17 678 Hispanic or Latino participants (30.6%), and 40 745 White participants (70.6%). Annualized incidence was 13.9% (95% CI, 13.3%-14.4%) for COVID-19 and 2.0% (95% CI, 1.8%-2.2%) for severe COVID-19. Factors associated with increased rates of COVID-19 included workplace exposure (high vs low: aHR, 1.35 [95% CI, 1.16-1.58]; medium vs low: aHR, 1.41 [95% CI, 1.21-1.65]; P < .001) and living condition risk (very high vs low risk: aHR, 1.41 [95% CI, 1.21-1.66]; medium vs low risk: aHR, 1.19 [95% CI, 1.08-1.32]; P < .001). Factors associated with decreased rates of COVID-19 included previous SARS-CoV-2 infection (aHR, 0.13 [95% CI, 0.09-0.19]; P < .001), age 65 years or older (aHR vs age <65 years, 0.57 [95% CI, 0.50-0.64]; P < .001) and Black or African American race (aHR vs White race, 0.78 [95% CI, 0.67-0.91]; P = .002). Factors associated with increased rates of severe COVID-19 included race (American Indian or Alaska Native vs White: aHR, 2.61 [95% CI, 1.85-3.69]; multiracial vs White: aHR, 2.19 [95% CI, 1.50-3.20]; P < .001), diabetes (aHR, 1.54 [95% CI, 1.14-2.08]; P = .005) and at least 2 comorbidities (aHR vs none, 1.39 [95% CI, 1.09-1.76]; P = .008). In analyses restricted to participants who contracted COVID-19, increased severe COVID-19 rates were associated with age 65 years or older (aHR vs <65 years, 1.75 [95% CI, 1.32-2.31]; P < .001), race (American Indian or Alaska Native vs White: aHR, 1.98 [95% CI, 1.38-2.83]; Black or African American vs White: aHR, 1.49 [95% CI, 1.03-2.14]; multiracial: aHR, 1.81 [95% CI, 1.21-2.69]; overall P = .001), body mass index (aHR per 1-unit increase, 1.03 [95% CI, 1.01-1.04]; P = .001), and diabetes (aHR, 1.85 [95% CI, 1.37-2.49]; P < .001). Previous SARS-CoV-2 infection was associated with decreased severe COVID-19 rates (aHR, 0.04 [95% CI, 0.01-0.14]; P < .001). Conclusions and Relevance In this secondary cross-protocol analysis of 4 randomized clinical trials, exposure and demographic factors had the strongest associations with outcomes; results could inform mitigation strategies for SARS-CoV-2 and viruses with comparable epidemiological characteristics.
Collapse
Affiliation(s)
- Deborah A. Theodore
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Angela R. Branche
- Department of Medicine, Infectious Disease Division, University of Rochester, Rochester, New York
| | - Lily Zhang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Daniel S. Graciaa
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Madhu Choudhary
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Raadhiya Osman
- Perinatal HIV Research Unit, Chris Hani Baragwanath Academic Hospital, Soweto, South Africa
| | - Tara M. Babu
- Department of Medicine, Division of Allergy & Infectious Diseases, University of Washington, Seattle
| | - Samuel T. Robinson
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Biostatistics, University of Washington, Seattle
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland
| | - Holly Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Biostatistics, University of Washington, Seattle
| | - James G. Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | | | - Paul Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham
| | - Glenda E. Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - Beatriz Grinsztejn
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Karen L. Kotloff
- Division of Infectious Disease and Tropical Pediatrics, Department of Pediatrics, University of Maryland School of Medicine, Baltimore
- Department of Medicine, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - Cynthia L. Gay
- Department of Medicine, Division of Infectious Diseases, UNC HIV Cure Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill
| | | | | | - Ian Hirsch
- AstraZeneca BioPharmaceuticals, Cambridge, United Kingdom
| | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | | | - Kathleen M. Neuzil
- Department of Medicine, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
| | - Ann R. Falsey
- Department of Medicine, Infectious Disease Division, University of Rochester, Rochester, New York
| | - Hana M. El Sahly
- Infectious Diseases Section, Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Magdalena E. Sobieszczyk
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Global Health, University of Washington, Seattle
| |
Collapse
|
398
|
Boyd MAA, Carey Hoppe A, Kelleher AD, Munier CML. T follicular helper cell responses to SARS-CoV-2 vaccination among healthy and immunocompromised adults. Immunol Cell Biol 2023; 101:504-513. [PMID: 36825370 PMCID: PMC10952589 DOI: 10.1111/imcb.12635] [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] [Received: 11/09/2022] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 02/25/2023]
Abstract
The worldwide rollout of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccinations in the last 2 years has produced a multitude of studies investigating T-cell responses in the peripheral blood and a limited number in secondary lymphoid tissues. As a key component to an effective immune response, vaccine-specific T follicular helper (Tfh) cells are localized in the draining lymph node (LN) and assist in the selection of highly specific B-cell clones for the production of neutralizing antibodies. While these cells have been noted in the blood as circulating Tfh (cTfh) cells, they are not often taken into consideration when examining effective CD4+ T-cell responses, particularly in immunocompromised groups. Furthermore, site-specific analyses in locations such as the LN have recently become an attractive area of investigation. This is mainly a result of improved sampling methods via ultrasound-guided fine-needle biopsy (FNB)/fine-needle aspiration (FNA), which are less invasive than LN excision and able to be performed longitudinally. While these studies have been undertaken in healthy individuals, data from immunocompromised groups are lacking. This review will focus on both Tfh and cTfh responses after SARS-CoV-2 vaccination in healthy and immunocompromised individuals. This area of investigation could identify key characteristics of a successful LN response required for the prevention of infection and viral clearance. This furthermore may highlight responses that could be fine-tuned to improve vaccine efficacy within immunocompromised groups that are at a risk of more severe disease.
Collapse
Affiliation(s)
| | - Alexandra Carey Hoppe
- Immunovirology and Pathogenesis ProgramThe Kirby InstituteUNSWSydneyNSW2052Australia
| | - Anthony D Kelleher
- Immunovirology and Pathogenesis ProgramThe Kirby InstituteUNSWSydneyNSW2052Australia
- St Vincent's HospitalSydneyNSW2010Australia
| | - C Mee Ling Munier
- Immunovirology and Pathogenesis ProgramThe Kirby InstituteUNSWSydneyNSW2052Australia
| |
Collapse
|
399
|
Hung Nguyen V, Boileau C, Bogdanov A, Sredl M, Bonafede M, Ducruet T, Chavers S, Rosen A, Martin D, Buck P, Esposito D, Van de Velde N, Mansi JA. Relative Effectiveness of BNT162b2, mRNA-1273, and Ad26.COV2.S Vaccines and Homologous Boosting in Preventing COVID-19 in Adults in the US. Open Forum Infect Dis 2023; 10:ofad288. [PMID: 37496607 PMCID: PMC10368199 DOI: 10.1093/ofid/ofad288] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/23/2023] [Indexed: 07/28/2023] Open
Abstract
Background Few head-to-head comparisons have been performed on the real-world effectiveness of coronavirus disease 2019 (COVID-19) booster vaccines. We evaluated the relative effectiveness (rVE) of a primary series of mRNA-1273 vs BNT162b2 and Ad26.COV2.S and a homologous mRNA booster against any medically attended, outpatient, and hospitalized COVID-19. Methods A data set linking primary care electronic medical records with medical claims data was used for this retrospective cohort study of US patients age ≥18 years vaccinated with a primary series between February and October 2021 (Part 1) and a homologous mRNA booster between October 2021 and January 2022 (Part 2). Adjusted hazard ratios (HRs) were derived from 1:1 matching adjusted across potential covariates. rVE was (1 - HRadjusted) × 100. Additional analysis was performed across regions and age groups. Results Following adjustment, Part 1 rVE for mRNA-1273 vs BNT162b2 was 23% (95% CI, 22%-25%), 23% (95% CI, 22%-25%), and 19% (95% CI, 14%-24%), while the rVE for mRNA-1273 vs Ad26.COV2.S was 50% (95% CI, 48%-51%), 50% (95% CI, 48%-52%), and 57% (95% CI, 53%-61%) against any medically attended, outpatient, and hospitalized COVID-19, respectively. The adjusted rVE in Part 2 for mRNA-1273 vs BNT162b2 was 14% (95% CI, 10%-18%), 13% (95% CI, 8%-17%), and 19% (95% CI, 1%-34%) against any medically attended, outpatient, and hospitalized COVID-19, respectively. rVE against medically attended COVID-19 was higher in adults age ≥65 years (35%; 95% CI, 24%-47%) than in those age 18-64 years (13%; 95% CI, 9%-17%) after the booster. Conclusions In this study, mRNA-1273 was more effective than BNT162b2 or Ad26.COV2.S following a primary series during the Delta-dominant period and more effective than BNT162b2 as a booster during the Omicron-dominant period.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Nicolas Van de Velde
- Correspondence: James A. Mansi, 5 Vaughn Drive, Princeton, NJ, 08540 (); or Nicolas Van de Velde, 5 Vaughn Drive, Princeton, NJ, 08540 ()
| | - James A Mansi
- Correspondence: James A. Mansi, 5 Vaughn Drive, Princeton, NJ, 08540 (); or Nicolas Van de Velde, 5 Vaughn Drive, Princeton, NJ, 08540 ()
| |
Collapse
|
400
|
van Balveren L, van Puijenbroek EP, Davidson L, van Hunsel F. A case series of bacillus Calmette-Guérin scar reactivation after administration of both mRNA and viral vector COVID-19 vaccines. Br J Clin Pharmacol 2023; 89:2113-2121. [PMID: 36717367 DOI: 10.1111/bcp.15678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/16/2022] [Accepted: 01/14/2023] [Indexed: 02/01/2023] Open
Abstract
AIM Reactivation of the scar resulting from intradermal injection of bacillus Calmette-Guérin (BCG) is a common specific reaction in Kawasaki's disease. It has also sporadically been associated with viral infections, multisystem inflammatory syndrome in children, influenza vaccination and mRNA COVID-19 vaccination. In this case series, characteristics of BCG scar reactivation after different COVID-19 vaccinations are presented and possible mechanisms are discussed. METHODS Data were collected from the spontaneous reporting system of the Netherlands Pharmacovigilance Centre Lareb. Descriptives were made for the case reports in which a BCG scar reactivation was detected. RESULTS Since the start of the COVID-19 vaccination campaign in January 2021, the Netherlands Pharmacovigilance Centre Lareb has received 22 case reports of BCG reactivation after vaccination with a COVID-19 vaccine. In 20 case reports, it concerned mRNA COVID-19 vaccines Moderna (14) and Pfizer (6). In two case reports, the viral vector COVID-19 vaccine AstraZeneca was administered. Erythema and pain were the most frequently reported symptoms and the size of the inflammation was between 1.5 and 5 cm. BCG scar reactivation occurred with a median time to onset of 2 days after the second or booster COVID-19 vaccination, whereas the median time to onset was 7 days after the first COVID-19 vaccination. None of the BCG scar reactivations were treated. CONCLUSIONS The exact mechanism of the occurrence of BCG scar reactivation remains unknown, but involvement of heat shock protein 65 is suggested. BCG scar reactivation is a nonserious, self-limiting reaction that can occur after vaccination with both mRNA and viral vector COVID-19 vaccines.
Collapse
Affiliation(s)
| | | | - Linda Davidson
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
| | - Florence van Hunsel
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
| |
Collapse
|