1
|
Agüero B, Berrios F, Pardo-Roa C, Ariyama N, Bennett B, Medina RA, Neira V. First detection of Omicron variant BA.4.1 lineage in dogs, Chile. Vet Q 2024; 44:1-10. [PMID: 38174799 PMCID: PMC10769545 DOI: 10.1080/01652176.2023.2298089] [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: 08/09/2023] [Accepted: 12/16/2023] [Indexed: 01/05/2024] Open
Abstract
SARS-CoV-2's rapid global spread caused the declaration of COVID-19 as a pandemic in March 2020. Alongside humans, domestic dogs and cats are also susceptible to infection. However, limited reports on pet infections in Chile prompted a comprehensive study to address this knowledge gap. Between March 2021 and March 2023, the study assessed 65 pets (26 dogs and 39 cats) from 33 COVID-19+ households alongside 700 nasal swabs from animals in households with unknown COVID-19 status. Using RT-PCR, nasal, fecal, and environmental samples were analyzed for the virus. In COVID-19+ households, 6.06% tested positive for SARS-CoV-2, belonging to 3 dogs, indicating human-to-pet transmission. Pets from households with unknown COVID-19 status tested negative for the virus. We obtained 2 SARS-CoV-2 genomes from animals, that belonged to Omicron BA.4.1 variant, marking the first report of pets infected with this lineage globally. Phylogenetic analysis showed these sequences clustered with human sequences collected in Chile during the same period when the BA.4.1 variant was prevalent in the country. The prevalence of SARS-CoV-2 in Chilean pets was relatively low, likely due to the country's high human vaccination rate. Our study highlights the importance of upholding and strengthening human vaccination strategies to mitigate the risk of interspecies transmission. It underscores the critical role of the One Health approach in addressing emerging zoonotic diseases, calling for further research on infection dynamics and risk factors for a comprehensive understanding.
Collapse
Affiliation(s)
- B. Agüero
- Programa de Doctorado en Ciencias Silvoagropecuarias y Veterinarias, Universidad de Chile, Santiago, Chile
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
| | - F. Berrios
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
| | - C. Pardo-Roa
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Child and Adolescent Health, School of Nursing, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - N. Ariyama
- Programa de Doctorado en Ciencias Silvoagropecuarias y Veterinarias, Universidad de Chile, Santiago, Chile
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
| | - B. Bennett
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
| | - RA. Medina
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - V. Neira
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
2
|
Lett MJ, Otte F, Hauser D, Schön J, Kipfer ET, Hoffmann D, Halwe NJ, Breithaupt A, Ulrich L, Britzke T, Kochmann J, Corleis B, Zhang Y, Urda L, Cmiljanovic V, Lang C, Beer M, Mittelholzer C, Klimkait T. High protection and transmission-blocking immunity elicited by single-cycle SARS-CoV-2 vaccine in hamsters. NPJ Vaccines 2024; 9:206. [PMID: 39472701 PMCID: PMC11522273 DOI: 10.1038/s41541-024-00992-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Vaccines have played a central role in combating the COVID-19 pandemic, but newly emerging SARS-CoV-2 variants are increasingly evading first-generation vaccine protection. To address this challenge, we designed "single-cycle infection SARS-CoV-2 viruses" (SCVs) that lack essential viral genes, possess distinctive immune-modulatory features, and exhibit an excellent safety profile in the Syrian hamster model. Animals intranasally vaccinated with an Envelope-gene-deleted vaccine candidate were fully protected against an autologous challenge with the SARS-CoV-2 virus through systemic and mucosal humoral immune responses. Additionally, the deletion of immune-downregulating viral genes in the vaccine construct prevented challenge virus transmission to contact animals. Moreover, vaccinated animals displayed neither tissue inflammation nor lung damage. Consequently, SCVs hold promising potential to induce potent protection against COVID-19, surpassing the immunity conferred by natural infection, as demonstrated in human immune cells.
Collapse
Affiliation(s)
- Martin Joseph Lett
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Fabian Otte
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - David Hauser
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Jacob Schön
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Enja Tatjana Kipfer
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Donata Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Nico J Halwe
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Angele Breithaupt
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Lorenz Ulrich
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Tobias Britzke
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Jana Kochmann
- Institute of Immunology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Björn Corleis
- Institute of Immunology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Yuepeng Zhang
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Lorena Urda
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Christopher Lang
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Christian Mittelholzer
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
- RocketVax AG, Basel, Switzerland
| | - Thomas Klimkait
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
3
|
Takahashi Y, Tanaka H, Taniguchi C, Ogata T. Efficiency of indirect protection of COVID-19 vaccination and interactions between indirect and direct protection on household transmission. Vaccine 2024; 42:126110. [PMID: 39019654 DOI: 10.1016/j.vaccine.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/20/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND There is little information on relationships between indirect and direct protection by COVID-19 vaccination on close contacts of the vaccinees. Here, we assessed effect modification of direct-indirect action influencing the protective effects of vaccination. METHODS Secondary attack rates (SARs) in household contacts (n = 2422) depending on vaccination status of the index cases (n = 1112) with known vaccination history during the delta variant-dominant period (August 2-November 2, 2021) in two public health jurisdictions were calculated using multivariable logistic regression analysis to assess indirect protection by COVID-19 vaccination as adjusted odds ratios (aORs) for SARs. The impact of the time of index case vaccination on indirect-direct protective effects was also assessed. FINDINGS Contacts of index cases receiving 2× COVID-19 vaccinations showed significantly lower SARs than contacts of unvaccinated index cases (aOR:0.48, 95 %CI = 0.32-0.74). Relative to contacts where neither index cases nor contacts themselves were vaccinated (0,0), those with (2,0), (0,2) and (2,2) had lower SARs (0.45, 95 %CI = 0.24-0.82, 0.24, 95 %CI = 0.17-0.032, 0.11, 95 %CI = 0.06-0.20, respectively. No significant interactions on the SARs regarding times of vaccination between index cases and household contacts were observed, indicating additive but not synergistic protection. INTERPRETATION The indirect protective effects of COVID-19 vaccination were attributed to an additive effect together with the direct effect on onward transmission in the household setting. These findings emphasize the importance of herd immunity by COVID-19 vaccination not only for unvaccinated but also vaccinated individuals.
Collapse
Affiliation(s)
- Yuki Takahashi
- Health and Medical General Affairs Division of Osaka Prefectural Government, Japan.
| | - Hideo Tanaka
- Neyagawa City Public Health Center, 28-3 Yasakacho, Neyagawa, Osaka, Japan.
| | - Chie Taniguchi
- College of Nursing, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, Japan.
| | - Tsuyoshi Ogata
- Itako Public Health Center of Ibaraki Prefectural Government, 1446-1 Osu, Itako, Ibaraki, Japan.
| |
Collapse
|
4
|
Tan ST, Rodríguez-Barraquer I, Kwan AT, Blumberg S, Park HJ, Hutchinson J, Leidner D, Lewnard JA, Sears D, Lo NC. Strength and durability of indirect protection against SARS-CoV-2 infection through vaccine and infection-acquired immunity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.23.24310889. [PMID: 39211889 PMCID: PMC11361209 DOI: 10.1101/2024.07.23.24310889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Early investigation revealed that COVID-19 vaccines confer indirect protection to fully susceptible and unvaccinated persons, defined as a reduced risk of SARS-CoV-2 infection among social contacts of vaccinated individuals. However, indirect protection from infection-acquired immunity and its comparative strength and durability to vaccine-derived indirect protection in the current epidemiologic context of high levels of vaccination, prior infection, and novel variants are not well characterized. Here, we show that both infection-acquired and vaccine-derived immunity independently yield indirect protection to close social contacts with key differences in their strength and waning. Analyzing anonymized data from a system-wide SARS-CoV-2 surveillance program of 177,319 residents across 35 California state prisons from December 2021 to December 2022 in a case-control design, we find that vaccine-derived indirect protection against Omicron SARS-CoV-2 infection is strongest within three months post-vaccination [30% (95% confidence interval: 20-38%)] with subsequent modest protection, whereas infection-acquired immunity provides 38% (24-50%) indirect protection to roommates for 6 months after SARS-CoV-2 infection, with moderate indirect protection persisting for over one year. Variant-targeted vaccines (bivalent formulation including Omicron subvariants BA.4/BA.5) confer strong indirect protection for at least three months [40% (3-63%)]. These results have important implications for understanding the long-term transmission dynamics of SARS-CoV-2 and can guide vaccine policy and public health measures, especially in high-risk environments such as prisons.
Collapse
|
5
|
Lin TY, Yen AMF, Chen SLS, Hsu CY, Lai CC, Luh DL, Yeh YP, Chen THH. Kinetics of Viral Shedding for Outbreak Surveillance of Emerging Infectious Diseases: Modeling Approach to SARS-CoV-2 Alpha and Omicron Infection. JMIR Public Health Surveill 2024; 10:e54861. [PMID: 39298261 PMCID: PMC11450350 DOI: 10.2196/54861] [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: 11/25/2023] [Revised: 07/09/2024] [Accepted: 07/21/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND Previous studies have highlighted the importance of viral shedding using cycle threshold (Ct) values obtained via reverse transcription polymerase chain reaction to understand the epidemic trajectories of SARS-CoV-2 infections. However, it is rare to elucidate the transition kinetics of Ct values from the asymptomatic or presymptomatic phase to the symptomatic phase before recovery using individual repeated Ct values. OBJECTIVE This study proposes a novel Ct-enshrined compartment model to provide a series of quantitative measures for delineating the full trajectories of the dynamics of viral load from infection until recovery. METHODS This Ct-enshrined compartment model was constructed by leveraging Ct-classified states within and between presymptomatic and symptomatic compartments before recovery or death among people with infections. A series of recovery indices were developed to assess the net kinetic movement of Ct-up toward and Ct-down off recovery. The model was applied to (1) a small-scale community-acquired Alpha variant outbreak under the "zero-COVID-19" policy without vaccines in May 2021 and (2) a large-scale community-acquired Omicron variant outbreak with high booster vaccination rates following the lifting of the "zero-COVID-19" policy in April 2022 in Taiwan. The model used Bayesian Markov chain Monte Carlo methods with the Metropolis-Hastings algorithm for parameter estimation. Sensitivity analyses were conducted by varying Ct cutoff values to assess the robustness of the model. RESULTS The kinetic indicators revealed a marked difference in viral shedding dynamics between the Alpha and Omicron variants. The Alpha variant exhibited slower viral shedding and lower recovery rates, but the Omicron variant demonstrated swifter viral shedding and higher recovery rates. Specifically, the Alpha variant showed gradual Ct-up transitions and moderate recovery rates, yielding a presymptomatic recovery index slightly higher than 1 (1.10), whereas the Omicron variant had remarkable Ct-up transitions and significantly higher asymptomatic recovery rates, resulting in a presymptomatic recovery index much higher than 1 (152.5). Sensitivity analysis confirmed the robustness of the chosen Ct values of 18 and 25 across different recovery phases. Regarding the impact of vaccination, individuals without booster vaccination had a 19% higher presymptomatic incidence rate compared to those with booster vaccination. Breakthrough infections in boosted individuals initially showed similar Ct-up transition rates but higher rates in later stages compared to nonboosted individuals. Overall, booster vaccination improved recovery rates, particularly during the symptomatic phase, although recovery rates for persistent asymptomatic infection were similar regardless of vaccination status once the Ct level exceeded 25. CONCLUSIONS The study provides new insights into dynamic Ct transitions, with the notable finding that Ct-up transitions toward recovery outpaced Ct-down and symptom-surfacing transitions during the presymptomatic phase. The Ct-up against Ct-down transition varies with variants and vaccination status. The proposed Ct-enshrined compartment model is useful for the surveillance of emerging infectious diseases in the future to prevent community-acquired outbreaks.
Collapse
Affiliation(s)
- Ting-Yu Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Amy Ming-Fang Yen
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sam Li-Sheng Chen
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chen-Yang Hsu
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Dachung Hospital, Miaoli, Taiwan
| | - Chao-Chih Lai
- Emergency Department of Taipei City Hospital, Ren-Ai Branch, Taipei, Taiwan
| | - Dih-Ling Luh
- Department of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Yen-Po Yeh
- Changhua Public Health Bureau, Chuang, Taiwan
| | - Tony Hsiu-Hsi Chen
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
6
|
Wilpstra CD, Morrell S, Mirza NA, Ralph JL. Consequences of COVID-19 Vaccine Hesitancy Among Healthcare Providers During the First 10 Months of Vaccine Availability: Scoping Review. Can J Nurs Res 2024; 56:204-224. [PMID: 38693882 PMCID: PMC11308270 DOI: 10.1177/08445621241251711] [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: 05/03/2024] Open
Abstract
BACKGROUND Throughout the COVID-19 pandemic, healthcare providers (HCPs)-including nurses-have played important roles in the vaccination effort. It is expected that COVID-19 vaccine hesitancy among HCPs has numerous consequences; however, the scope of these consequences and their impacts on providers, patients, and the broader healthcare system remained unclear. PURPOSE To identify existing and emerging evidence to understand the state of knowledge of the consequences of COVID-19 vaccine hesitancy among HCPs. METHODS A scoping review was completed based upon the JBI scoping review methodology. The databases searched included OVID Medline, EBSCOhost CINAHL, ProQuest Nursing and Allied Health Source, ProQuest APA PsycInfo, and ProQuest Dissertations and Theses. The final literature search was completed on June 2, 2022. Studies were screened and retrieved based on predefined inclusion and exclusion criteria using Covidence reference management software. Data extraction followed criteria recommended in the JBI scoping review framework with additional relevant variables identified by the authors. RESULTS A total of 33 sources were included in the review. Consequences of HCP COVID-19 vaccine hesitancy were grouped under three themes and seven subthemes. Consequences affecting HCPs included health-related, psychosocial, and employment-related consequences. Consequences affecting patients pertained to COVID-19 vaccination communication and COVID-19 vaccination practices of HCPs. Consequences to the healthcare system involved consequences to coworkers and employment/attendance/staffing-related consequences. CONCLUSIONS Healthcare provider COVID-19 vaccine hesitancy was found to have numerous consequences. By understanding the scope and extent of these consequences, healthcare leaders, researchers, and HCPs can work together to protect providers, patients, and healthcare systems.
Collapse
Affiliation(s)
| | - Sherry Morrell
- Faculty of Nursing, University of Windsor, Windsor, Canada
| | | | - Jody L. Ralph
- Faculty of Nursing, University of Windsor, Windsor, Canada
| |
Collapse
|
7
|
Edwards CT, Karunakaran KA, Garcia E, Beutler N, Gagne M, Golden N, Aoued H, Pellegrini KL, Burnett MR, Honeycutt CC, Lapp SA, Ton T, Lin MC, Metz A, Bombin A, Goff K, Scheuermann SE, Wilkes A, Wood JS, Ehnert S, Weissman S, Curran EH, Roy M, Dessasau E, Paiardini M, Upadhyay AA, Moore I, Maness NJ, Douek DC, Piantadosi A, Andrabi R, Rogers TR, Burton DR, Bosinger SE. Passive infusion of an S2-Stem broadly neutralizing antibody protects against SARS-CoV-2 infection and lower airway inflammation in rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605768. [PMID: 39109178 PMCID: PMC11302620 DOI: 10.1101/2024.07.30.605768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The continued evolution of SARS-CoV-2 variants capable of subverting vaccine and infection-induced immunity suggests the advantage of a broadly protective vaccine against betacoronaviruses (β-CoVs). Recent studies have isolated monoclonal antibodies (mAbs) from SARS-CoV-2 recovered-vaccinated donors capable of neutralizing many variants of SARS-CoV-2 and other β-CoVs. Many of these mAbs target the conserved S2 stem region of the SARS-CoV-2 spike protein, rather the receptor binding domain contained within S1 primarily targeted by current SARS-CoV-2 vaccines. One of these S2-directed mAbs, CC40.8, has demonstrated protective efficacy in small animal models against SARS-CoV-2 challenge. As the next step in the pre-clinical testing of S2-directed antibodies as a strategy to protect from SARS-CoV-2 infection, we evaluated the in vivo efficacy of CC40.8 in a clinically relevant non-human primate model by conducting passive antibody transfer to rhesus macaques (RM) followed by SARS-CoV-2 challenge. CC40.8 mAb was intravenously infused at 10mg/kg, 1mg/kg, or 0.1 mg/kg into groups (n=6) of RM, alongside one group that received a control antibody (PGT121). Viral loads in the lower airway were significantly reduced in animals receiving higher doses of CC40.8. We observed a significant reduction in inflammatory cytokines and macrophages within the lower airway of animals infused with 10mg/kg and 1mg/kg doses of CC40.8. Viral genome sequencing demonstrated a lack of escape mutations in the CC40.8 epitope. Collectively, these data demonstrate the protective efficiency of broadly neutralizing S2-targeting antibodies against SARS-CoV-2 infection within the lower airway while providing critical preclinical work necessary for the development of pan-β-CoV vaccines.
Collapse
Affiliation(s)
- Christopher T. Edwards
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kirti A. Karunakaran
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, TN 37235, USA
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, Minnesota 55356, USA
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Matthew Gagne
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, LA, USA
| | - Hadj Aoued
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kathryn L. Pellegrini
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Matthew R. Burnett
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Cole Honeycutt
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stacey A. Lapp
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Thang Ton
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Mark C. Lin
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Amanda Metz
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Andrei Bombin
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Amelia Wilkes
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jennifer S. Wood
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Stephanie Ehnert
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Stacey Weissman
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Elizabeth H. Curran
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Melissa Roy
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Evan Dessasau
- Division of Histology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Amit A. Upadhyay
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Ian Moore
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Daniel C. Douek
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anne Piantadosi
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Thomas R. Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
8
|
Bratti Frank Terre BR, de Oliveira Toso BRG, Reis LF, Johann JA. Analysis of public policies to combat COVID-19 in the state of Paraná, Brazil. Front Public Health 2024; 12:1384561. [PMID: 39086801 PMCID: PMC11288802 DOI: 10.3389/fpubh.2024.1384561] [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: 02/09/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction The COVID-19 pandemic had a great impact on several public sectors worldwide, requiring the implementation of public policies in an organized way to contain and control the disease. Thus, this study aimed to analyze public policies to face the COVID-19 pandemic in the State of Paraná, Brazil. Methods This was a cross-sectional, retrospective, quantitative survey carried out with data from March 2020 to March 2022 in the twenty-two municipalities that host the local health regions. Data collection was documentary, carried out from the municipal Portal da Transparência website, which is dedicated to making public all expenditures, and epidemiological bulletins released by the Health Department of the state of Paraná. The variables analyzed were New Cases and Deaths, Mortality and Lethality Coefficient, Incidence Rate, Vaccination Coverage, Number of Hospital and ICU beds exclusive to COVID-19, Settled Expenses destined for COVID-19 and coping measures, namely, the Declaration of Public Health Emergency, Curfew, Mandatory use of masks, Businesses closure, Teleworking of risk groups, and Suspension of activities with crowds and of classes. After collection, data underwent descriptive analysis, and subsequently, the correlation of variables was analyzed using the Spearman test. Multiple linear regression was applied using the variable selection method called best subset selection (BSS). The dependent variables listed were incidence rate, new cases and new deaths. Results The results showed that coping measures, as well as the application of resources for the pandemic, were implemented heterogeneously in the municipalities studied, and the progression of the disease, the distribution of beds and vaccination occurred unevenly and may be a reflection of the limited Brazilian national governance. An important correlation was observed between the incidence rate and new deaths with vaccination coverage. In addition, the regression model showed that measures such as mandatory use of masks, closure of shops, suspension of classes, and curfew showed important correlations with the variables incidence rate, cases, and new deaths. Discussion The study highlighted the importance of carrying out a robust analysis of public policies to face emergencies of global importance so that government entities are prepared for future crises of great repercussions, such as the COVID-19 pandemic.
Collapse
Affiliation(s)
- Bruna Regina Bratti Frank Terre
- Centro de Ciências Biológicas e da Saúde, Programa de Pós-graduação Stricto Sensu em Biociências e Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
| | - Beatriz Rosana Gonçalves de Oliveira Toso
- Centro de Ciências Biológicas e da Saúde, Programa de Pós-graduação Stricto Sensu em Biociências e Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
| | - Luiz Fernando Reis
- Centro de Ciências Biológicas e da Saúde, Programa de Pós-graduação Stricto Sensu em Biociências e Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
| | - Jerry Adriani Johann
- Pró-Reitoria de Pesquisa de Pós-graduação, Programa de Pós-graduação Stricto Sensu em Engenharia Agrícola, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
| |
Collapse
|
9
|
Herbert C, Antar AA, Broach J, Wright C, Stamegna P, Luzuriaga K, Hafer N, McManus DD, Manabe YC, Soni A. Relationship between acute SARS-CoV-2 viral clearance with Long COVID Symptoms: a cohort study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.04.24309953. [PMID: 39006428 PMCID: PMC11245049 DOI: 10.1101/2024.07.04.24309953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Introduction The relationship between SARS-CoV-2 viral dynamics during acute infection and the development of long COVID is largely unknown. Methods A total of 7361 asymptomatic community-dwelling people enrolled in the Test Us at Home parent study between October 2021 and February 2022. Participants self-collected anterior nasal swabs for SARS-CoV-2 RT-PCR testing every 24-48 hours for 10-14 days, regardless of symptom or infection status. Participants who had no history of COVID-19 at enrollment and who were subsequently found to have ≥1 positive SARS-CoV-2 RT-PCR test during the parent study were recontacted in August 2023 and asked whether they had experienced long COVID, defined as the development of new symptoms lasting 3 months or longer following SARS-CoV-2 infection. Participant's cycle threshold values were converted into viral loads, and slopes of viral clearance were modeled using post-nadir viral loads. Using a log binomial model with the modeled slopes as the exposure, we calculated the relative risk of subsequently developing long COVID with 1-2 symptoms, 3-4 symptoms, or 5+ symptoms, adjusting for age, number of symptoms, and SARS-CoV-2 variant. Adjusted relative risk (aRR) of individual long COVID symptoms based on viral clearance was also calculated. Results 172 participants were eligible for analyses, and 59 (34.3%) reported experiencing long COVID. The risk of long COVID with 3-4 symptoms and 5+ symptoms increased by 2.44 times (aRR: 2.44; 95% CI: 0.88-6.82) and 4.97 times (aRR: 4.97; 95% CI: 1.90-13.0) per viral load slope-unit increase, respectively. Participants who developed long COVID had significantly longer times from peak viral load to viral clearance during acute disease than those who never developed long COVID (8.65 [95% CI: 8.28-9.01] vs. 10.0 [95% CI: 9.25-10.8]). The slope of viral clearance was significantly positively associated with long COVID symptoms of fatigue (aRR: 2.86; 95% CI: 1.22-6.69), brain fog (aRR: 4.94; 95% CI: 2.21-11.0), shortness of breath (aRR: 5.05; 95% CI: 1.24-20.6), and gastrointestinal symptoms (aRR: 5.46; 95% CI: 1.54-19.3). Discussion We observed that longer time from peak viral load to viral RNA clearance during acute COVID-19 was associated with an increased risk of developing long COVID. Further, slower clearance rates were associated with greater number of symptoms of long COVID. These findings suggest that early viral-host dynamics are mechanistically important in the subsequent development of long COVID.
Collapse
Affiliation(s)
- Carly Herbert
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- University of Massachusetts Center for Clinical and Translational Science, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Annukka A.R. Antar
- Division of Infectious Disease, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Broach
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Colton Wright
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Pamela Stamegna
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Katherine Luzuriaga
- University of Massachusetts Center for Clinical and Translational Science, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Nathaniel Hafer
- University of Massachusetts Center for Clinical and Translational Science, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - David D McManus
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Division of Health System Science, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Division of Cardiology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yukari C Manabe
- Division of Infectious Disease, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Apurv Soni
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Division of Health System Science, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
10
|
Harris CE, Vaidya V, Lan Z, Klompas M, Rhee C, Baden LR, Baker MA. SARS-CoV-2 Polymerase Chain Reaction Cycle Threshold Trends in Patients Who Are Immunocompromised and Implications for Isolation Precautions. Open Forum Infect Dis 2024; 11:ofae367. [PMID: 39077053 PMCID: PMC11285146 DOI: 10.1093/ofid/ofae367] [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: 03/11/2024] [Accepted: 06/26/2024] [Indexed: 07/31/2024] Open
Abstract
Among 495 patients who were immunocompromised and tested positive for SARS-CoV-2, polymerase chain reaction cycle thresholds remained <33 beyond 20 days more frequently in patients with hematologic malignancies, particularly those receiving B-cell-depleting or Bruton tyrosine kinase inhibitor therapy, as compared with those with solid organ malignancy (26% vs 5%).
Collapse
Affiliation(s)
- Courtney E Harris
- Division of Infectious Disease, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Vineeta Vaidya
- Infection Control Department, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zhou Lan
- Center for Clinical Investigation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Klompas
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Chanu Rhee
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lindsey R Baden
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Meghan A Baker
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Benoit TJ, Kim Y, Deng Y, Li Z, Harding L, Wiegand R, Deng X, Jones JM, Ronaldo I, Clarke KEN. Association Between Social Vulnerability and SARS-CoV-2 Seroprevalence in Specimens Collected From Commercial Laboratories, United States, September 2021-February 2022. Public Health Rep 2024; 139:501-511. [PMID: 38357883 DOI: 10.1177/00333549231223140] [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: 02/16/2024] Open
Abstract
OBJECTIVE We conducted a national US study of SARS-CoV-2 seroprevalence by Social Vulnerability Index (SVI) that included pediatric data and compared the Delta and Omicron periods during the COVID-19 pandemic. The objective of the current study was to assess the association between SVI and seroprevalence of infection-induced SARS-CoV-2 antibodies by period (Delta vs Omicron) and age group. METHODS We used results of infection-induced SARS-CoV-2 antibody assays of clinical sera specimens (N = 406 469) from 50 US states from September 2021 through February 2022 to estimate seroprevalence overall and by county SVI tercile. Bivariate analyses and multilevel logistic regression models assessed the association of seropositivity with SVI and its themes by age group (0-17, ≥18 y) and period (Delta: September-November 2021; Omicron: December 2021-February 2022). RESULTS Aggregate infection-induced SARS-CoV-2 antibody seroprevalence increased at all 3 SVI levels; it ranged from 25.8% to 33.5% in September 2021 and from 53.1% to 63.5% in February 2022. Of the 4 SVI themes, socioeconomic status had the strongest association with seroprevalence. During the Delta period, we found significantly more infections per reported case among people living in a county with high SVI (odds ratio [OR] = 2.76; 95% CI, 2.31-3.21) than in a county with low SVI (OR = 1.65; 95% CI, 1.33-1.97); we found no significant difference during the Omicron period. Otherwise, findings were consistent across subanalyses by age group and period. CONCLUSIONS Among both children and adults, and during both the Delta and Omicron periods, counties with high SVI had significantly higher SARS-CoV-2 antibody seroprevalence than counties with low SVI did. These disparities reinforce SVI's value in identifying communities that need tailored prevention efforts during public health emergencies and resources to recover from their effects.
Collapse
Affiliation(s)
- Tina J Benoit
- COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | - Zheng Li
- Office of Capacity Development and Applied Prevention Science, Agency for Toxic Substances and Disease Registry, Atlanta, GA, USA
| | | | - Ryan Wiegand
- COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Jefferson M Jones
- COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Kristie E N Clarke
- COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
12
|
Venkat H, Yaglom HD, Hecht G, Goedderz A, Ely JL, Sprenkle M, Martins T, Jasso-Selles D, Lemmer D, Gesimondo J, Ruberto I, Komatsu K, Engelthaler DM. Investigation of SARS-CoV-2 Infection among Companion Animals in Households with Confirmed Human COVID-19 Cases. Pathogens 2024; 13:466. [PMID: 38921764 PMCID: PMC11206992 DOI: 10.3390/pathogens13060466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/17/2024] [Accepted: 05/31/2024] [Indexed: 06/27/2024] Open
Abstract
We aimed to characterize SARS-CoV-2 infection in companion animals living in households with COVID-19-positive people and understand the dynamics surrounding how these animals become infected. Public health investigators contacted households with at least one confirmed, symptomatic person with COVID-19 for study recruitment. Blood, nasal, and rectal swab specimens were collected from pet dogs and cats and a questionnaire was completed. Specimens were tested for SARS-CoV-2 by RT-PCR, and for neutralizing antibodies; genomic sequencing was performed on viral-positive samples. A total of 36.4% of 110 pets enrolled had evidence of infection with SARS-CoV-2. Pets were more likely to test positive if the pet was immunocompromised, and if more than one person in the home was positive for COVID-19. Among 12 multi-pet households where at least one pet was positive, 10 had at least one other pet test positive. Whole-genome sequencing revealed the genomes of viral lineages circulating in the community during the time of sample collection. Our findings suggest a high likelihood of viral transmission in households with multiple pets and when pets had very close interactions with symptomatic humans. Further surveillance studies are needed to characterize how new variants impact animals and to understand opportunities for infection and spillover in susceptible species.
Collapse
Affiliation(s)
- Heather Venkat
- Arizona Department of Health Services, Phoenix, AZ 85007, USA; (G.H.); (I.R.); (K.K.)
- Career Epidemiology Field Officer Program, Center for Preparedness and Response, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Hayley D. Yaglom
- Translational Genomics Research Institute, Pathogen and Microbiome Division, Flagstaff, AZ 86005, USA (D.L.); (D.M.E.)
| | - Gavriella Hecht
- Arizona Department of Health Services, Phoenix, AZ 85007, USA; (G.H.); (I.R.); (K.K.)
| | - Andrew Goedderz
- Translational Genomics Research Institute, Pathogen and Microbiome Division, Flagstaff, AZ 86005, USA (D.L.); (D.M.E.)
| | - Jennifer L. Ely
- Translational Genomics Research Institute, Pathogen and Microbiome Division, Flagstaff, AZ 86005, USA (D.L.); (D.M.E.)
| | - Michael Sprenkle
- Translational Genomics Research Institute, Pathogen and Microbiome Division, Flagstaff, AZ 86005, USA (D.L.); (D.M.E.)
| | - Taylor Martins
- Arizona Department of Health Services, Phoenix, AZ 85007, USA; (G.H.); (I.R.); (K.K.)
| | - Daniel Jasso-Selles
- Translational Genomics Research Institute, Pathogen and Microbiome Division, Flagstaff, AZ 86005, USA (D.L.); (D.M.E.)
| | - Darrin Lemmer
- Translational Genomics Research Institute, Pathogen and Microbiome Division, Flagstaff, AZ 86005, USA (D.L.); (D.M.E.)
| | | | - Irene Ruberto
- Arizona Department of Health Services, Phoenix, AZ 85007, USA; (G.H.); (I.R.); (K.K.)
| | - Kenneth Komatsu
- Arizona Department of Health Services, Phoenix, AZ 85007, USA; (G.H.); (I.R.); (K.K.)
| | - David M. Engelthaler
- Translational Genomics Research Institute, Pathogen and Microbiome Division, Flagstaff, AZ 86005, USA (D.L.); (D.M.E.)
| |
Collapse
|
13
|
Khezri M, McKnight C, Weng CA, Kimball S, Des Jarlais D. COVID-19 vaccination uptake and determinants of booster vaccination among persons who inject drugs in New York City. PLoS One 2024; 19:e0303394. [PMID: 38743729 PMCID: PMC11093290 DOI: 10.1371/journal.pone.0303394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Persons who inject drugs (PWID) may be unengaged with healthcare services and face an elevated risk of severe morbidity and mortality associated with COVID-19 due to chronic diseases and structural inequities. However, data on COVID-19 vaccine uptake, particularly booster vaccination, among PWID are limited. We examined COVID-19 vaccine uptake and factors associated with booster vaccination among PWID in New York City (NYC). METHODS We recruited PWID using respondent-driven sampling from October 2021 to November 2023 in a survey that included HIV and SARS-CoV-2 antibodies testing. The questionnaire included demographics, COVID-19 vaccination and attitudes, and drug use behaviors. RESULTS Of 436 PWID, 80% received at least one COVID-19 vaccine dose. Among individuals who received at least one COVID-19 vaccine dose, 95% were fully vaccinated. After excluding participants recruited before booster authorization for general adults started in NYC, and those who had never received an initial vaccination, 41% reported having received a COVID-19 booster vaccine dose. COVID-19 booster vaccination was significantly associated with having a high school diploma or GED (adjusted odds ratio (aOR) 1.93; 95% confidence interval (CI) 1.09, 3.48), ever received the hepatitis A/B vaccine (aOR 2.23; 95% CI 1.27, 3.96), main drug use other than heroin/speedball, fentanyl and stimulants (aOR 14.4; 95% CI 2.32, 280), number of non-fatal overdoses (aOR 0.35; 95% CI 0.16, 0.70), and mean vaccination attitude score (aOR 0.94; 95% CI 0.89, 0.98). CONCLUSIONS We found a suboptimal level of COVID-19 booster vaccination among PWID, which was consistent with the rates observed in the general population in NYC and the U.S. Community-based interventions are needed to improve COVID-19 booster vaccination access and uptake among PWID. Attitudes towards vaccination were significant predictors of both primary and booster vaccination uptake. Outreach efforts focusing on improving attitudes towards vaccination and educational programs are essential for reducing hesitancy and increasing booster vaccination uptake among PWID.
Collapse
Affiliation(s)
- Mehrdad Khezri
- Department of Epidemiology, School of Global Public Health, New York University, New York, NY, United States of America
- HIV/STI Surveillance Research Center, and WHO Collaborating Center for HIV Surveillance, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Courtney McKnight
- Department of Epidemiology, School of Global Public Health, New York University, New York, NY, United States of America
- Center for Drug Use and HIV/HCV Research, New York, NY, United States of America
| | - Chenziheng Allen Weng
- Department of Epidemiology, School of Global Public Health, New York University, New York, NY, United States of America
| | - Sarah Kimball
- Department of Epidemiology, School of Global Public Health, New York University, New York, NY, United States of America
| | - Don Des Jarlais
- Department of Epidemiology, School of Global Public Health, New York University, New York, NY, United States of America
- Center for Drug Use and HIV/HCV Research, New York, NY, United States of America
| |
Collapse
|
14
|
Oordt-Speets AM, Spinardi JR, Mendoza CF, Yang J, del Carmen Morales G, Kyaw MH. Duration of SARS-CoV-2 shedding: A systematic review. J Glob Health 2024; 14:05005. [PMID: 38547496 PMCID: PMC10978056 DOI: 10.7189/jogh.14.05005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024] Open
Abstract
Background Positive viral severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cultures indicate shedding of infectious virus and corresponding transmission risk of coronavirus disease 2019 (COVID-19). The research question of this systematic review was: Is there a discernible pattern in the timing of SARS-CoV-2 virus isolation, and what is the proportion of positive and negative results for isolation of SARS-CoV-2 virus with viral culture relative to the onset of clinical symptoms or the day of diagnosis, as indicated by longitudinal studies? Methods We systematically searched PubMed and Embase from inception to 16 February 2023 for English-language studies with serial viral culture testing within symptomatic or asymptomatic SARS-CoV-2 infected persons during the post-vaccination period. Outcomes of interest were the daily culture status per study and the overall daily culture positivity rate of SARS-CoV-2. We critically appraised the selected studies using the Newcastle-Ottawa quality assessment scale. Results We included 14 viral shedding studies in this systematic review. Positive viral SARS-CoV-2 cultures were detected in samples ranging from 4 days before to 18 days after symptom onset. The daily culture SARS-CoV-2 positivity rate since symptom onset or diagnosis showed a steep decline between day 5 and 9, starting with a peak ranging from 44% to 50% on days -1 to 5, decreasing to 28% on day 7 and 11% on day 9, and finally ranging between 0% and 8% on days 10-17. Conclusions Viral shedding peaked within 5 days since symptom onset or diagnosis and the culture positivity rate rapidly declined hereafter. This systematic review provides an overview of current evidence on the daily SARS-CoV-2 culture positivity rates during the post-vaccination period. These findings could be used to estimate the effectiveness of public health control measures, including treatment and preventive strategies, to reduce the spread of COVID-19.
Collapse
Affiliation(s)
| | - Julia R Spinardi
- Vaccine Medical Affairs, Emerging Markets, Pfizer Inc., Itapevi, Brazil
| | | | - Jingyan Yang
- Global Value and Access, Pfizer Inc., New York, USA
| | | | - Moe H Kyaw
- Vaccine Scientific Affairs, Emerging Markets, Pfizer Inc., New York, USA
| |
Collapse
|
15
|
Ghosh A, Kanta P, Ramola M, Mohindra R, Goyal K, Kishore R, Suri V, Lakshmi PVM, Chauhan C, Sharma M, Rakshit P, Ponnusamy K, Dikid T, Singh MP. Rapid Decline of SARS-CoV-2 Viral Load in Single vs. Double-Dose (Short-Interval <6 Weeks) ChAdOx nCoV-19 Vaccinated Health-Care Workers. Curr Microbiol 2024; 81:95. [PMID: 38353761 DOI: 10.1007/s00284-023-03603-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 12/26/2023] [Indexed: 02/16/2024]
Abstract
The present work was carried out during the emergence of Delta Variant of Concern (VoC) and aimed to study the change in SARS CoV-2 viral load in Covishield vaccinated asymptomatic/mildly symptomatic health-care workers (HCWs) to find out the optimum isolation period. The SARS CoV-2 viral load was carried out in sequential samples of 55 eligible HCWs which included unvaccinated (UnV; n = 11), single-dose vaccinated (SDV, n = 20) and double-dose vaccinated [DDV, n = 24; short-interval (<6 weeks)] subjects. The mean load of envelope (E) gene on day 5 in SDV [0.42 × 105 copies/reaction] was significantly lower as compared to DDV [6.3 × 105 copies/reaction, P = 0.005] and UnV [6.6 × 105 copies/reaction, P = 0.001] groups. The rate of decline of SARS CoV-2 viral load in the initial 5 days of PCR positivity was significantly higher in SDV as compared to that in DDV (Mean log decline 0.39 vs. 0.19; P < 0.001). This was possibly due to interference of adenoviral immunity of first dose of adenovirus-vectored vaccine in double-dose vaccinated HCWs who had received vaccines within a shorter interval (<6 weeks).
Collapse
Affiliation(s)
- Arnab Ghosh
- Department of Virology, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Poonam Kanta
- Department of Virology, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Manisha Ramola
- Department of Virology, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Ritin Mohindra
- Department of Internal Medicine, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Kapil Goyal
- Department of Virology, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Roop Kishore
- Department of Internal Medicine, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Vikas Suri
- Department of Internal Medicine, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - P V M Lakshmi
- Department of Community Medicine and School of Public Health, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Chanderkanta Chauhan
- Department of Community Medicine and School of Public Health, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Manisha Sharma
- Department of Community Medicine and School of Public Health, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Partha Rakshit
- National Centre for Disease Control (NCDC), New Delhi, India
| | | | - Tanzin Dikid
- National Centre for Disease Control (NCDC), New Delhi, India
| | - Mini P Singh
- Department of Virology, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India.
| |
Collapse
|
16
|
Kampf G. Does the COVID-19 Vaccination Reduce the Risk to Transmit SARS-CoV-2 to Others? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1457:247-264. [PMID: 39283431 DOI: 10.1007/978-3-031-61939-7_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
It has been assumed that the COVID-19 vaccination reduces the risk of transmission to others. Results during the delta predominance show that the viral load in the vaccinated population is not consistently lower compared to the unvaccinated, and during the omicron predominance, the viral load was even somewhat higher. Levels of infectious SARS-CoV-2 were partly lower in the vaccinated population. Viral loads were mostly lower in re-infections compared to breakthrough infections. Viral clearance including the detection of infectious virus has mostly been described to be faster in the vaccinated population suggesting a shorter duration as a possible source for transmission. The epidemiological relevance of this finding remains uncertain. Approximately half of the transmission studies found lower secondary attack rates from the fully vaccinated population, but the results are probably best explained by the vaccination status of the contact population. Public health data from the UK show that the number of COVID-19 cases is higher among the fully vaccinated and boosted population who might be possible sources, in contrast to lower case numbers within the first three months among the vaccinated obtained in phase 3 trials on symptomatic cases. Overall, there is no convincing evidence that the COVID-19 vaccination significantly reduces the risk to transmit SARS-CoV-2 to others.
Collapse
Affiliation(s)
- Günter Kampf
- University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, 17475, Greifswald, Germany.
| |
Collapse
|
17
|
Port JR, Yinda CK, Riopelle JC, Weishampel ZA, Saturday TA, Avanzato VA, Schulz JE, Holbrook MG, Barbian K, Perry-Gottschalk R, Haddock E, Martens C, Shaia CI, Lambe T, Gilbert SC, van Doremalen N, Munster VJ. Infection- or AZD1222 vaccine-mediated immunity reduces SARS-CoV-2 transmission but increases Omicron competitiveness in hamsters. Nat Commun 2023; 14:6592. [PMID: 37852960 PMCID: PMC10584863 DOI: 10.1038/s41467-023-42346-8] [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: 01/19/2023] [Accepted: 10/06/2023] [Indexed: 10/20/2023] Open
Abstract
Limited data is available on the effect of vaccination and previous virus exposure on the nature of SARS-CoV-2 transmission and immune-pressure on variants. To understand the impact of pre-existing immunity on SARS-CoV-2 airborne transmission efficiency, we perform a transmission chain experiment using naïve, intranasally or intramuscularly AZD1222 vaccinated, and previously infected hamsters. A clear gradient in transmission efficacy is observed: Transmission in hamsters vaccinated via the intramuscular route was reduced over three airborne chains (approx. 60%) compared to naïve animals, whereas transmission in previously infected hamsters and those vaccinated via the intranasal route was reduced by 80%. We also find that the Delta B.1.617.2 variant outcompeted Omicron B.1.1.529 after dual infection within and between hosts in naïve, vaccinated, and previously infected transmission chains, yet an increase in Omicron B.1.1.529 competitiveness is observed in groups with pre-existing immunity against Delta B.1.617.2. This correlates with an increase in the strength of the humoral response against Delta B.1.617.2, with the strongest response seen in previously infected animals. These data highlight the continuous need to improve vaccination strategies and address the additional evolutionary pressure pre-existing immunity may exert on SARS-CoV-2.
Collapse
Affiliation(s)
- Julia R Port
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Claude Kwe Yinda
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jade C Riopelle
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Zachary A Weishampel
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Taylor A Saturday
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Victoria A Avanzato
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jonathan E Schulz
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Myndi G Holbrook
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kent Barbian
- Genomics Research Section, Research Technologies Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Rose Perry-Gottschalk
- Rocky Mountain Visual and Medical Arts Unit, Research Technologies Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Elaine Haddock
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Craig Martens
- Genomics Research Section, Research Technologies Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Carl I Shaia
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Teresa Lambe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Chinese Academy of Medical Science Oxford Institute; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Sarah C Gilbert
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Neeltje van Doremalen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Vincent J Munster
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
18
|
Alfano V, Capasso S, Limosani M. On the determinants of anti-COVID restriction and anti-vaccine movements: the case of IoApro in Italy. Sci Rep 2023; 13:16784. [PMID: 37798271 PMCID: PMC10556032 DOI: 10.1038/s41598-023-42133-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 09/05/2023] [Indexed: 10/07/2023] Open
Abstract
Following restrictions to control the spread of COVID-19, and subsequent vaccination campaigns, sentiments against such policies were quick to arise. While individual-level determinants that led to such attitudes have drawn much attention, there are also reasons to believe that the macro context in which these movements arose may contribute to their evolution. In this study, exploiting data on business activities which supported a major Italian anti-restriction and anti-vaccine movement, IoApro, using quantitative analysis that employs both a fractional response probit and logit model and a beta regression model, we investigate the relationship between socio-economic characteristics, institutional quality, and the flourishing of this movement. Our results suggest a U-shaped relationship between income and the proliferation of the movement, meaning that support for these movements increases the greater the degree of economic decline. Our results further indicate that the share of the population between 40 and 60 years old is positively related to support for such movements, as is institutional corruption.
Collapse
Affiliation(s)
- Vincenzo Alfano
- DiSEGIM, University of Napoli Parthenope, Naples, Italy.
- Center for Economic Studies - CES-Ifo, Munich, Germany.
| | - Salvatore Capasso
- Department of Human and Social Sciences, Italian National Research Council, Rome, Italy
- University of Napoli Parthenope, Naples, Italy
- CSEF, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
19
|
Hong P, Li C, Tian X, Che D. Factors influencing viral shedding time in non-severe paediatric infection with the SARS-CoV-2: a single-centre retrospective study. BMJ Paediatr Open 2023; 7:e001991. [PMID: 37827803 PMCID: PMC10582883 DOI: 10.1136/bmjpo-2023-001991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/28/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND The aim of this study was to determine the factors influencing viral shedding time (VST) in non-severe paediatric infection with SARS-CoV-2). METHODS We conducted a retrospective analysis of data from 240 non-severe paediatric infection with the SARS-CoV-2. Multivariate Cox regression analysis was used to identify independent predictors associated with VST. RESULTS Two hundred and forty patients were included in the study. The median duration of VST was 10 days (IQR, 8-13 days). Compared with patients aged <1 year, children aged 6-12 years (adjusted HR (aHR): 1.849; 95% CI 1.031 to 3.315) and >12 years (aHR: 2.180; 95% CI 1.071 to 4.439) had shorter VST. Non-leucopenia patients (aHR: 1.431; 95% CI 1.005 to 2.038) also had a lower VST. DISCUSSION The results of this study show that children aged <1 year and children with leucopenia had longer SARS-CoV-2 VST. These factors should be taken into account when developing policies for the isolation of patients with COVID-19.
Collapse
Affiliation(s)
- Ping Hong
- Pediatrics department, Shanghai Changhai Hospital, Shanghai, China
- International Medical Centre, Shanghai Children's Hospital, School of medicine, Shanghai Jiaotong University, Shanghai, China
| | - Chengmei Li
- International Medical Centre, Shanghai Children's Hospital, School of medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xianmin Tian
- Department of Clinical Laboratory, Shanghai Children's Hospital, Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Datian Che
- International Medical Centre, Shanghai Children's Hospital, School of medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
20
|
Chang TH, Wu CH, Chen PY, Ho SY, Chung MY, Sheng WH, Lu CY, Yen TY, Chen JM, Lee PI, Tang HJ, Ho CH, Chang LY, Chen YC, Huang LM. Viral dynamics of SARS-CoV-2 Omicron infections in a previously low COVID-19 prevalence region: Effects of vaccination status, antiviral agents, and age. J Formos Med Assoc 2023; 122:872-879. [PMID: 37179128 PMCID: PMC10151453 DOI: 10.1016/j.jfma.2023.04.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/28/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND In Taiwan, the prevalence of COVID-19 was low before April 2022. The low SARS-CoV-2 seroprevalence in the population of Taiwan provides an opportunity for comparison with fewer confounding factors than other populations globally. Cycle threshold (Ct) value is an easily accessible method for modeling SARS-CoV-2 dynamics. In this study, we used clinical samples collected from hospitalized patients to explore the Ct value dynamics of the Omicron variant infection. METHODS From Jan 2022 to May 2022, we retrospectively included hospitalized patients tested positive by nasopharyngeal SARS-CoV-2 PCR. We categorized the test-positive subjects into different groups according to age, vaccination status, and use of antiviral agents. To investigate the nonlinear relationship between symptom onset days and Ct value, a fractional polynomial model was applied to draw a regression line. RESULTS We collected 1718 SARS-CoV-2 viral samples from 812 individuals. The Ct values of unvaccinated individuals were lower than those of vaccinated persons from Day 4 to Day 10 after symptom onset. The Ct value increased more rapidly in those individuals with antiviral drug treatment from Day 2 to Day 7. In elderly individuals, the Ct values increased slowly from Day 5 to Day 10, and the increasing trend was unique compared with that in children and adults. CONCLUSION Our study demonstrated the primary viral infection dynamics of the Omicron variant in hospitalized patients. Vaccination significantly affected viral dynamics, and antiviral agents modified viral dynamics irrespective of vaccination status. In elderly individuals, viral clearance is slower than that in adults and children.
Collapse
Affiliation(s)
- Tu-Hsuan Chang
- Department of Pediatrics, Chi Mei Medical Center, Tainan, Taiwan
| | - Chi-Hsien Wu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, Cardinal Tien Hospital, Taipei, Taiwan
| | - Po-Yu Chen
- Department of Nursing, Min-Hwei College of Health Care Management, Tainan, Taiwan
| | - Shu-Yuan Ho
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Yi Chung
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wang-Huei Sheng
- Division of Infectious Diseases, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chun-Yi Lu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Ting-Yu Yen
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Jong-Min Chen
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ping-Ing Lee
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Hung-Jen Tang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Chung-Han Ho
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan; Department of Information Management, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Luan-Yin Chang
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.
| | - Yee-Chun Chen
- Division of Infectious Diseases, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Li-Min Huang
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
21
|
Maeda M, Murata F, Fukuda H. Effect of COVID-19 vaccination on household transmission of SARS-CoV-2 in the Omicron era: The Vaccine Effectiveness, Networking, and Universal Safety (VENUS) study. Int J Infect Dis 2023; 134:200-206. [PMID: 37356650 PMCID: PMC10289267 DOI: 10.1016/j.ijid.2023.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/19/2023] [Accepted: 06/19/2023] [Indexed: 06/27/2023] Open
Abstract
OBJECTIVES To evaluate the effectiveness of vaccination on reducing household transmission of SARS-CoV-2 among common household types in Japan during the Omicron variant wave. METHODS This retrospective study was conducted using vaccination records, COVID-19 infection data, and resident registry data from two Japanese municipalities. Households that experienced their first COVID-19 case between January and April 2022 were categorized into two groups according to the presence/absence of children aged ≤11 years. We constructed multivariable logistic regression models with generalized estimating equations to calculate the odds ratios (ORs) and 95% confidence intervals for household transmission according to the vaccination statuses of primary cases and household contacts. RESULTS We analyzed 7326 households with 17,586 contacts. In all households, the OR for household transmission was <0.6 (P <0.001) when the primary case and/or contact were vaccinated. In households with children aged ≤11 years, the OR was 0.71 (P <0.001) when only the contact was vaccinated. In households with all members aged ≥12 years, the OR was <0.5 (P <0.001) when the primary case and/or contact were vaccinated. CONCLUSION COVID-19 vaccination effectively reduced household transmission in Japan during the Omicron variant wave.
Collapse
Affiliation(s)
- Megumi Maeda
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumiko Murata
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Haruhisa Fukuda
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
22
|
Alpuche-Lazcano SP, Stuible M, Akache B, Tran A, Kelly J, Hrapovic S, Robotham A, Haqqani A, Star A, Renner TM, Blouin J, Maltais JS, Cass B, Cui K, Cho JY, Wang X, Zoubchenok D, Dudani R, Duque D, McCluskie MJ, Durocher Y. Preclinical evaluation of manufacturable SARS-CoV-2 spike virus-like particles produced in Chinese Hamster Ovary cells. COMMUNICATIONS MEDICINE 2023; 3:116. [PMID: 37612423 PMCID: PMC10447459 DOI: 10.1038/s43856-023-00340-7] [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: 03/16/2023] [Accepted: 07/25/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND As the COVID-19 pandemic continues to evolve, novel vaccines need to be developed that are readily manufacturable and provide clinical efficacy against emerging SARS-CoV-2 variants. Virus-like particles (VLPs) presenting the spike antigen at their surface offer remarkable benefits over other vaccine antigen formats; however, current SARS-CoV-2 VLP vaccines candidates in clinical development suffer from challenges including low volumetric productivity, poor spike antigen density, expression platform-driven divergent protein glycosylation and complex upstream/downstream processing requirements. Despite their extensive use for therapeutic protein manufacturing and proven ability to produce enveloped VLPs, Chinese Hamster Ovary (CHO) cells are rarely used for the commercial production of VLP-based vaccines. METHODS Using CHO cells, we aimed to produce VLPs displaying the full-length SARS-CoV-2 spike. Affinity chromatography was used to capture VLPs released in the culture medium from engineered CHO cells expressing spike. The structure, protein content, and glycosylation of spikes in VLPs were characterized by several biochemical and biophysical methods. In vivo, the generation of neutralizing antibodies and protection against SARS-CoV-2 infection was tested in mouse and hamster models. RESULTS We demonstrate that spike overexpression in CHO cells is sufficient by itself to generate high VLP titers. These VLPs are evocative of the native virus but with at least three-fold higher spike density. In vivo, purified VLPs elicit strong humoral and cellular immunity at nanogram dose levels which grant protection against SARS-CoV-2 infection. CONCLUSIONS Our results show that CHO cells are amenable to efficient manufacturing of high titers of a potently immunogenic spike protein-based VLP vaccine antigen.
Collapse
Affiliation(s)
- Sergio P Alpuche-Lazcano
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Bassel Akache
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Anh Tran
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - John Kelly
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Dr, Ottawa, ON, K1A 0R6, Canada
| | - Sabahudin Hrapovic
- Aquatic and Crop Resources Development Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Anna Robotham
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Dr, Ottawa, ON, K1A 0R6, Canada
| | - Arsalan Haqqani
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Dr, Ottawa, ON, K1A 0R6, Canada
| | - Alexandra Star
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Dr, Ottawa, ON, K1A 0R6, Canada
| | - Tyler M Renner
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Julie Blouin
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Jean-Sébastien Maltais
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Brian Cass
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Kai Cui
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB, T6G 2M9, Canada
| | - Jae-Young Cho
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB, T6G 2M9, Canada
| | - Xinyu Wang
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB, T6G 2M9, Canada
| | - Daria Zoubchenok
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Renu Dudani
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Diana Duque
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Michael J McCluskie
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada.
| |
Collapse
|
23
|
Plachouras D, Kacelnik O, Rodríguez-Baño J, Birgand G, Borg MA, Kristensen B, Kubele J, Lyytikäinen O, Presterl E, Reilly J, Voss A, Zingg W, Suetens C, Monnet DL. Revisiting the personal protective equipment components of transmission-based precautions for the prevention of COVID-19 and other respiratory virus infections in healthcare. Euro Surveill 2023; 28:2200718. [PMID: 37561052 PMCID: PMC10416576 DOI: 10.2807/1560-7917.es.2023.28.32.2200718] [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: 08/30/2022] [Accepted: 05/10/2023] [Indexed: 08/11/2023] Open
Abstract
The COVID-19 pandemic highlighted some potential limitations of transmission-based precautions. The distinction between transmission through large droplets vs aerosols, which have been fundamental concepts guiding infection control measures, has been questioned, leading to considerable variation in expert recommendations on transmission-based precautions for COVID-19. Furthermore, the application of elements of contact precautions, such as the use of gloves and gowns, is based on low-quality and inconclusive evidence and may have unintended consequences, such as increased incidence of healthcare-associated infections and spread of multidrug-resistant organisms. These observations indicate a need for high-quality studies to address the knowledge gaps and a need to revisit the theoretical background regarding various modes of transmission and the definitions of terms related to transmission. Further, we should examine the implications these definitions have on the following components of transmission-based precautions: (i) respiratory protection, (ii) use of gloves and gowns for the prevention of respiratory virus infections, (iii) aerosol-generating procedures and (iv) universal masking in healthcare settings as a control measure especially during seasonal epidemics. Such a review would ensure that transmission-based precautions are consistent and rationally based on available evidence, which would facilitate decision-making, guidance development and training, as well as their application in practice.
Collapse
Affiliation(s)
| | | | - Jesús Rodríguez-Baño
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases and Microbiology division, Hospital Universitario Virgen Macarena and Department of Medicine, University of Seville/CSIC, Biomedicine Institute of Seville, Seville, Spain
| | - Gabriel Birgand
- Health Protection Research Unit, Healthcare Associated Infection and Antimicrobial Resistance, Imperial College London, London, United Kingdom
- Regional Centre for Infection Prevention and Control, Region of Pays de la Loire, Nantes, France
| | - Michael A Borg
- Infection Control Department, Mater Dei Hospital, Msida, Malta
| | | | - Jan Kubele
- Clinical Microbiology and ATB centre, Na Homolce Hospital, Prague, Czechia
| | | | - Elisabeth Presterl
- Department for Hospital Epidemiology and Infection Control, Medical University of Vienna, Vienna, Austria
| | - Jacqui Reilly
- Research Centre for Health, Glasgow Caledonian University, Glasgow, United Kingdom
| | - Andreas Voss
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, the Netherlands
| | - Walter Zingg
- Charité Universitätsmedizin, Berlin, Germany
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Carl Suetens
- European Centre for Disease Prevention and Control, Stockholm, Sweden
| | | |
Collapse
|
24
|
Roche R, Odeh NH, Andar AU, Tulapurkar ME, Roche JA. Protection against Severe Illness versus Immunity-Redefining Vaccine Effectiveness in the Aftermath of COVID-19. Microorganisms 2023; 11:1963. [PMID: 37630523 PMCID: PMC10459411 DOI: 10.3390/microorganisms11081963] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/03/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Anti-SARS-CoV-2 vaccines have played a pivotal role in reducing the risk of developing severe illness from COVID-19, thus helping end the COVID-19 global public health emergency after more than three years. Intriguingly, as SARS-CoV-2 variants emerged, individuals who were fully vaccinated did get infected in high numbers, and viral loads in vaccinated individuals were as high as those in the unvaccinated. However, even with high viral loads, vaccinated individuals were significantly less likely to develop severe illness; this begs the question as to whether the main effect of anti-SARS-CoV-2 vaccines is to confer protection against severe illness or immunity against infection. The answer to this question is consequential, not only to the understanding of how anti-SARS-CoV-2 vaccines work, but also to public health efforts against existing and novel pathogens. In this review, we argue that immune system sensitization-desensitization rather than sterilizing immunity may explain vaccine-mediated protection against severe COVID-19 illness even when the SARS-CoV-2 viral load is high. Through the lessons learned from COVID-19, we make the case that in the disease's aftermath, public health agencies must revisit healthcare policies, including redefining the term "vaccine effectiveness."
Collapse
Affiliation(s)
- Renuka Roche
- Occupational Therapy Program, School of Health Sciences, College of Health and Human Services, Eastern Michigan University, Ypsilanti, MI 48197, USA;
| | - Nouha H. Odeh
- Ph.D. Program in Immunology and Microbiology, Department of Biochemistry, Microbiology & Immunology, School of Medicine, Wayne State University, Detroit, MI 48201, USA;
| | - Abhay U. Andar
- Baltimore County, Translational Life Science Technology, University of Maryland, Rockville, MD 20850, USA;
| | - Mohan E. Tulapurkar
- Division of Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joseph A. Roche
- Physical Therapy Program, Department of Health Care Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
25
|
Landelle C, Birgand G, Price JR, Mutters NT, Morgan DJ, Lucet JC, Kerneis S, Zingg W. Considerations for de-escalating universal masking in healthcare centers. ANTIMICROBIAL STEWARDSHIP & HEALTHCARE EPIDEMIOLOGY : ASHE 2023; 3:e128. [PMID: 37592969 PMCID: PMC10428150 DOI: 10.1017/ash.2023.200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/10/2023] [Accepted: 05/10/2023] [Indexed: 08/19/2023]
Abstract
Three years after the beginning of the COVID-19 pandemic, better knowledge on the transmission of respiratory viral infections (RVI) including the contribution of asymptomatic infections encouraged most healthcare centers to implement universal masking. The evolution of the SARS-CoV-2 epidemiology and improved immunization of the population call for the infection and prevention control community to revisit the masking strategy in healthcare. In this narrative review, we consider factors for de-escalating universal masking in healthcare centers, addressing compliance with the mask policy, local epidemiology, the level of protection provided by medical face masks, the consequences of absenteeism and presenteeism, as well as logistics, costs, and ecological impact. Most current national and international guidelines for mask use are based on the level of community transmission of SARS-CoV-2. Actions are now required to refine future recommendations, such as establishing a list of the most relevant RVI to consider, implement reliable local RVI surveillance, and define thresholds for activating masking strategies. Considering the epidemiological context (measured via sentinel networks or wastewater analysis), and, if not available, considering a time period (winter season) may guide to three gradual levels of masking: (i) standard and transmission-based precautions and respiratory etiquette, (ii) systematic face mask wearing when in direct contact with patients, and (iii) universal masking. Cost-effectiveness analysis of the different strategies is warranted in the coming years. Masking is just one element to be considered along with other preventive measures such as staff and patient immunization, and efficient ventilation.
Collapse
Affiliation(s)
- Caroline Landelle
- University of Grenoble Alpes, CNRS, UMR 5525, Grenoble INP, CHU Grenoble Alpes, Infection Prevention and Control Unit, 38000 Grenoble, France
| | - Gabriel Birgand
- National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at Imperial College London, London, UK
- Regional Center for Infection Prevention and Control Pays de la Loire, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | | | - Nico T. Mutters
- Institute for Hygiene and Public Health, University Hospital Bonn, Bonn, Germany
| | - Daniel J. Morgan
- University of Maryland School of Medicine, Baltimore, MD, USA
- VA Maryland Healthcare System, Baltimore, MD, USA
| | - Jean-Christophe Lucet
- Infection Control Unit, Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Solen Kerneis
- Infection Control Unit, Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Walter Zingg
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
26
|
Li F, Deng J, Xie C, Wang G, Xu M, Wu C, Li J, Zhong Y. The differences in virus shedding time between the Delta variant and original SARS-CoV-2 infected patients. Front Public Health 2023; 11:1132643. [PMID: 37559731 PMCID: PMC10408444 DOI: 10.3389/fpubh.2023.1132643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 07/06/2023] [Indexed: 08/11/2023] Open
Abstract
Background The worldwide epidemic of Coronavirus Disease 2019 (COVID-19) has evolved into multiple variants. The Delta variant is known for its ability to spread and replicate, while data are limited about the virus shedding time in patients infected by the Delta variant. Methods 56 Delta variant and 56 original SARS-CoV-2 infected patients from Hunan, China, matched according to age and gender divided into two groups and compared the baseline characteristics and laboratory findings with appropriate statistical methods. Results Patients infected with the Delta variant had significantly fewer symptoms of fever (p < 0.001), fatigue (p = 0.004), anorexia (p < 0.001), shortness of breath (p = 0.004), diarrhea (p = 0.006), positive pneumonia rate of chest CT (p = 0.019) and chest CT ground glass opacities (p = 0.004) than those of patients with the original SARS-CoV-2. Patients of the Delta variant group had a significantly longer virus shedding time [41.5 (31.5, 46.75) vs. 18.5 (13, 25.75), p < 0.001] compared with the original SARS-CoV-2 group. The correlation analyses between the virus shedding time and clinical or laboratory parameters showed that the virus shedding time was positively related to the viral strain, serum creatinine and creatine kinase isoenzyme, while negatively correlated with lymphocyte count, total bilirubin and low-density lipoprotein. Finally, the viral strain and lymphocyte count were thought of as the independent risk factors of the virus shedding time demonstrated by multiple linear regression. Conclusion COVID-19 patients infected with the Delta variant exhibited fewer gastrointestinal symptoms and prolonged virus shedding time than those infected with the original SARS-CoV-2. Delta variant and fewer lymphocyte were correlated with prolonged virus shedding time.
Collapse
Affiliation(s)
- Fanglin Li
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiayi Deng
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Canbin Xie
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Guyi Wang
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Min Xu
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chenfang Wu
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jinxiu Li
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yanjun Zhong
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
27
|
Frutos AM, Kuan G, Lopez R, Ojeda S, Shotwell A, Sanchez N, Saborio S, Plazaola M, Barilla C, Kenah E, Balmaseda A, Gordon A. Infection-Induced Immunity Is Associated With Protection Against Severe Acute Respiratory Syndrome Coronavirus 2 Infection and Decreased Infectivity. Clin Infect Dis 2023; 76:2126-2133. [PMID: 36774538 PMCID: PMC10273383 DOI: 10.1093/cid/ciad074] [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: 10/10/2022] [Revised: 01/24/2023] [Accepted: 02/08/2023] [Indexed: 02/13/2023] Open
Abstract
BACKGROUND The impact of infection-induced immunity on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) transmission has not been well established. Here we estimate the effects of prior infection induced immunity in adults and children on SARS-CoV-2 transmission in households. METHODS We conducted a household cohort study from March 2020-November 2022 in Managua, Nicaragua; following a housheold SARS-CoV-2 infection, household members are closely monitored for infection. We estimate the association of time period, age, symptoms, and prior infection with secondary attack risk. RESULTS Overall, transmission occurred in 70.2% of households, 40.9% of household contacts were infected, and the secondary attack risk ranged from 8.1% to 13.9% depending on the time period. Symptomatic infected individuals were more infectious (rate ratio [RR] 21.2, 95% confidence interval [CI]: 7.4-60.7) and participants with a prior infection were half as likely to be infected compared to naïve individuals (RR 0.52, 95% CI:.38-.70). In models stratified by age, prior infection was associated with decreased infectivity in adults and adolescents (secondary attack risk [SAR] 12.3, 95% CI: 10.3, 14.8 vs 17.5, 95% CI: 14.8, 20.7). However, although young children were less likely to transmit, neither prior infection nor symptom presentation was associated with infectivity. During the Omicron era, infection-induced immunity remained protective against infection. CONCLUSIONS Infection-induced immunity is associated with decreased infectivity for adults and adolescents. Although young children are less infectious, prior infection and asymptomatic presentation did not reduce their infectivity as was seen in adults. As SARS-CoV-2 transitions to endemicity, children may become more important in transmission dynamics.
Collapse
Affiliation(s)
- Aaron M Frutos
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Guillermina Kuan
- Health Center Sócrates Flores Vivas, Ministry of Health, Managua, Nicaragua
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Roger Lopez
- Sustainable Sciences Institute, Managua, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Sergio Ojeda
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Abigail Shotwell
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Nery Sanchez
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Saira Saborio
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
- Sustainable Sciences Institute, Managua, Nicaragua
| | | | | | - Eben Kenah
- Biostatistics Division, College of Public Health, The Ohio State University, Columbus, Ohio, USA
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
28
|
Corchis-Scott R, Geng Q, Al Riahi AM, Labak A, Podadera A, Ng KKS, Porter LA, Tong Y, Dixon JC, Menard SL, Seth R, McKay RM. Actionable wastewater surveillance: application to a university residence hall during the transition between Delta and Omicron resurgences of COVID-19. Front Public Health 2023; 11:1139423. [PMID: 37265515 PMCID: PMC10230041 DOI: 10.3389/fpubh.2023.1139423] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Wastewater surveillance has gained traction during the COVID-19 pandemic as an effective and non-biased means to track community infection. While most surveillance relies on samples collected at municipal wastewater treatment plants, surveillance is more actionable when samples are collected "upstream" where mitigation of transmission is tractable. This report describes the results of wastewater surveillance for SARS-CoV-2 at residence halls on a university campus aimed at preventing outbreak escalation by mitigating community spread. Another goal was to estimate fecal shedding rates of SARS-CoV-2 in a non-clinical setting. Passive sampling devices were deployed in sewer laterals originating from residence halls at a frequency of twice weekly during fall 2021 as the Delta variant of concern continued to circulate across North America. A positive detection as part of routine sampling in late November 2021 triggered daily monitoring and further isolated the signal to a single wing of one residence hall. Detection of SARS-CoV-2 within the wastewater over a period of 3 consecutive days led to a coordinated rapid antigen testing campaign targeting the residence hall occupants and the identification and isolation of infected individuals. With knowledge of the number of individuals testing positive for COVID-19, fecal shedding rates were estimated to range from 3.70 log10 gc ‧ g feces-1 to 5.94 log10 gc ‧ g feces-1. These results reinforce the efficacy of wastewater surveillance as an early indicator of infection in congregate living settings. Detections can trigger public health measures ranging from enhanced communications to targeted coordinated testing and quarantine.
Collapse
Affiliation(s)
- Ryland Corchis-Scott
- Great Lakes Institute for Environmental Research, University of Windsor, Windsor, ON, Canada
| | - Qiudi Geng
- Great Lakes Institute for Environmental Research, University of Windsor, Windsor, ON, Canada
| | - Abdul Monem Al Riahi
- Great Lakes Institute for Environmental Research, University of Windsor, Windsor, ON, Canada
| | - Amr Labak
- Great Lakes Institute for Environmental Research, University of Windsor, Windsor, ON, Canada
| | - Ana Podadera
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, Canada
| | - Kenneth K. S. Ng
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, Canada
| | - Lisa A. Porter
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, Canada
| | - Yufeng Tong
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, Canada
| | - Jess C. Dixon
- Department of Kinesiology, University of Windsor, Windsor, ON, Canada
| | | | - Rajesh Seth
- Civil and Environmental Engineering, University of Windsor, Windsor, ON, Canada
| | - R. Michael McKay
- Great Lakes Institute for Environmental Research, University of Windsor, Windsor, ON, Canada
| |
Collapse
|
29
|
Braeye T, Catteau L, Brondeel R, van Loenhout JAF, Proesmans K, Cornelissen L, Van Oyen H, Stouten V, Hubin P, Billuart M, Djiena A, Mahieu R, Hammami N, Van Cauteren D, Wyndham-Thomas C. Vaccine effectiveness against transmission of alpha, delta and omicron SARS-COV-2-infection, Belgian contact tracing, 2021-2022. Vaccine 2023; 41:3292-3300. [PMID: 37085456 PMCID: PMC10073587 DOI: 10.1016/j.vaccine.2023.03.069] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023]
Abstract
OBJECTIVES Vaccine effectiveness against transmission (VET) of SARS-CoV-2-infection can be estimated from secondary attack rates observed during contact tracing. We estimated VET, the vaccine-effect on infectiousness of the index case and susceptibility of the high-risk exposure contact (HREC). METHODS We fitted RT-PCR-test results from HREC to immunity status (vaccine schedule, prior infection, time since last immunity-conferring event), age, sex, calendar week of sampling, household, background positivity rate and dominant VOC using a multilevel Bayesian regression-model. We included Belgian data collected between January 2021 and January 2022. RESULTS For primary BNT162b2-vaccination we estimated initial VET at 96% (95%CI 95-97) against Alpha, 87% (95%CI 84-88) against Delta and 31% (95%CI 25-37) against Omicron. Initial VET of booster-vaccination (mRNA primary and booster-vaccination) was 87% (95%CI 86-89) against Delta and 68% (95%CI 65-70) against Omicron. The VET-estimate against Delta and Omicron decreased to 71% (95%CI 64-78) and 55% (95%CI 46-62) respectively, 150-200 days after booster-vaccination. Hybrid immunity, defined as vaccination and documented prior infection, was associated with durable and higher or comparable (by number of antigen exposures) protection against transmission. CONCLUSIONS While we observed VOC-specific immune-escape, especially by Omicron, and waning over time since immunization, vaccination remained associated with a reduced risk of SARS-CoV-2-transmission.
Collapse
Affiliation(s)
- Toon Braeye
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium.
| | - Lucy Catteau
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Ruben Brondeel
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Joris A F van Loenhout
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Kristiaan Proesmans
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Laura Cornelissen
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Herman Van Oyen
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium; Department of Public Health and Primary Care, Ghent University, Corneel Heymanslaan 10, 9000 Gent, Belgium
| | - Veerle Stouten
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Pierre Hubin
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Matthieu Billuart
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Achille Djiena
- Agence pour une Vie de Qualité, Rue de la Rivelaine 11, 6061 Charleroi, Belgium
| | - Romain Mahieu
- Common Community Commission Brussels, Rue Belliard 71/1, 1040 Brussels, Belgium
| | - Naima Hammami
- Agency for Care and Health, Infection Prevention and Control, Flemish Community, Koningin Maria Hendrikaplein 70 bus 55, 9000 Gent, Belgium
| | - Dieter Van Cauteren
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Chloé Wyndham-Thomas
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| |
Collapse
|
30
|
Van Egeren D, Stoddard M, White LF, Hochberg NS, Rogers MS, Zetter B, Joseph-McCarthy D, Chakravarty A. Vaccines Alone Cannot Slow the Evolution of SARS-CoV-2. Vaccines (Basel) 2023; 11:853. [PMID: 37112765 PMCID: PMC10143044 DOI: 10.3390/vaccines11040853] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/11/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
The rapid emergence of immune-evading viral variants of SARS-CoV-2 calls into question the practicality of a vaccine-only public-health strategy for managing the ongoing COVID-19 pandemic. It has been suggested that widespread vaccination is necessary to prevent the emergence of future immune-evading mutants. Here, we examined that proposition using stochastic computational models of viral transmission and mutation. Specifically, we looked at the likelihood of emergence of immune escape variants requiring multiple mutations and the impact of vaccination on this process. Our results suggest that the transmission rate of intermediate SARS-CoV-2 mutants will impact the rate at which novel immune-evading variants appear. While vaccination can lower the rate at which new variants appear, other interventions that reduce transmission can also have the same effect. Crucially, relying solely on widespread and repeated vaccination (vaccinating the entire population multiple times a year) is not sufficient to prevent the emergence of novel immune-evading strains, if transmission rates remain high within the population. Thus, vaccines alone are incapable of slowing the pace of evolution of immune evasion, and vaccinal protection against severe and fatal outcomes for COVID-19 patients is therefore not assured.
Collapse
Affiliation(s)
- Debra Van Egeren
- Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
- New York Genome Center, New York, NY 10013, USA
| | | | - Laura F. White
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Natasha S. Hochberg
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Michael S. Rogers
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Bruce Zetter
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | | | | |
Collapse
|
31
|
Ong DSY, de Man P, Verhagen T, Doejaaren G, Dallinga MA, Alibux E, Janssen ML, Wils EJ. Airborne virus shedding of the alpha, delta, omicron SARS-CoV-2 variants and influenza virus in hospitalized patients. J Med Virol 2023; 95:e28748. [PMID: 37185846 DOI: 10.1002/jmv.28748] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/22/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023]
Abstract
Airborne transmission is an important transmission route for the spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Epidemiological data indicate that certain SARS-CoV-2 variants, like the omicron variant, are associated with higher transmissibility. We compared virus detection in air samples between hospitalized patients infected with different SARS-CoV-2 variants or influenza virus. The study was performed during three separate time periods in which subsequently the alpha, delta, and omicron SARS-CoV-2 variants were predominant. In total, 79 patients with coronavirus disease 2019 (COVID-19) and 22 patients with influenza A virus infection were included. Collected air samples were positive in 55% of patients infected with the omicron variant in comparison to 15% of those infected with the delta variant (p < 0.01). In multivariable analysis, the SARS-CoV-2 omicron BA.1/BA.2 variant (as compared to the delta variant) and the viral load in nasopharynx were both independently associated with air sample positivity, but the alpha variant and COVID-19 vaccination were not. The proportion of positive air samples patients infected with the influenza A virus was 18%. In conclusion, the higher air sample positivity rate of the omicron variant compared to previous SARS-CoV-2 variants may partially explain the higher transmission rates seen in epidemiological trends.
Collapse
Affiliation(s)
- David S Y Ong
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter de Man
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Tim Verhagen
- Department of Intensive Care, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Gerda Doejaaren
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Marloes A Dallinga
- Department of Pulmonary Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Esmee Alibux
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Matthijs L Janssen
- Department of Intensive Care, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Intensive Care, Erasmus MC, Rotterdam, The Netherlands
| | - Evert-Jan Wils
- Department of Intensive Care, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
- Department of Intensive Care, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
32
|
Ye J, Shao X, Yang Y, Zhu F. Predicting the negative conversion time of nonsevere COVID-19 patients using machine learning methods. J Med Virol 2023; 95:e28747. [PMID: 37185847 DOI: 10.1002/jmv.28747] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/10/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023]
Abstract
Based on the patient's clinical characteristics and laboratory indicators, different machine-learning methods were used to develop models for predicting the negative conversion time of nonsevere coronavirus disease 2019 (COVID-19) patients. A retrospective analysis was performed on 376 nonsevere COVID-19 patients admitted to Wuxi Fifth People's Hospital from May 2, 2022, to May 14, 2022. The patients were divided into training set (n = 309) and test set (n = 67). The clinical features and laboratory parameters of the patients were collected. In the training set, the least absolute shrinkage and selection operator (LASSO) was used to select predictive features and train six machine learning models: multiple linear regression (MLR), K-Nearest Neighbors Regression (KNNR), random forest regression (RFR), support vector machine regression (SVR), XGBoost regression (XGBR), and multilayer perceptron regression (MLPR). Seven best predictive features selected by LASSO included: age, gender, vaccination status, IgG, lymphocyte ratio, monocyte ratio, and lymphocyte count. The predictive performance of the models in the test set was MLPR > SVR > MLR > KNNR > XGBR > RFR, and MLPR had the strongest generalization performance, which is significantly better than SVR and MLR. In the MLPR model, vaccination status, IgG, lymphocyte count, and lymphocyte ratio were protective factors for negative conversion time; male gender, age, and monocyte ratio were risk factors. The top three features with the highest weights were vaccination status, gender, and IgG. Machine learning methods (especially MLPR) can effectively predict the negative conversion time of non-severe COVID-19 patients. It can help to rationally allocate limited medical resources and prevent disease transmission, especially during the Omicron pandemic.
Collapse
Affiliation(s)
- Jiru Ye
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaonan Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging of Soochow University, Changzhou Clinical Medical Center, Changzhou, China
| | - Yong Yang
- Department of Pediatrics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Feng Zhu
- Department of Respiratory and Critical Care Medicine, Affiliated Wuxi Fifth Hospital of Jiangnan University, Wuxi Fifth People's Hospital, Wuxi, China
| |
Collapse
|
33
|
Comparison of culture-competent virus shedding duration of SARS-CoV-2 omicron variant in regard to vaccination status: a prospective cohort study. Vaccine 2023; 41:2769-2772. [PMID: 37003909 PMCID: PMC10040344 DOI: 10.1016/j.vaccine.2023.03.044] [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: 11/22/2022] [Revised: 02/20/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
Previous studies have shown that fully vaccinated patients with SARS-CoV-2 Delta variants has shorter viable viral shedding period compared to unvaccinated or partially vaccinated patients. However, data about effects of vaccination against the viable viral shedding period in patients with SARS-CoV-2 Omicron variants were limited. We compared the viable viral shedding period of SARS-CoV-2 omicron variant regard to vaccination status. Saliva samples were obtained daily from patients with SARS-CoV-2 Omicron variant, and genomic assessments and virus culture was performed to those samples. We found no difference in viable viral shedding period between fully vaccinated and not or partially vaccinated, nor between 1st boostered vs non-boostered patients with SARS-CoV-2 Omicron variant.
Collapse
|
34
|
Marking U, Havervall S, Norin NG, Bladh O, Christ W, Gordon M, Ng H, Blom K, Phillipson M, Mangsbo S, Alm JJ, Smed-Sörensen A, Nilsson P, Hober S, Åberg M, Klingström J, Thålin C. Correlates of protection and viral load trajectories in omicron breakthrough infections in triple vaccinated healthcare workers. Nat Commun 2023; 14:1577. [PMID: 36949041 PMCID: PMC10031702 DOI: 10.1038/s41467-023-36984-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 02/27/2023] [Indexed: 03/24/2023] Open
Abstract
Vaccination offers protection against severe COVID-19 caused by SARS-CoV-2 omicron but is less effective against infection. Characteristics such as serum antibody titer correlation to protection, viral abundance and clearance of omicron infection in vaccinated individuals are scarce. We present a 4-week twice-weekly SARS-CoV-2 qPCR screening in 368 triple vaccinated healthcare workers. Spike-specific IgG levels, neutralization titers and mucosal spike-specific IgA-levels were determined at study start and qPCR-positive participants were sampled repeatedly for two weeks. 81 (cumulative incidence 22%) BA.1, BA.1.1 and BA.2 infections were detected. High serum antibody titers are shown to be protective against infection (p < 0.01), linked to reduced viral load (p < 0.01) and time to viral clearance (p < 0.05). Pre-omicron SARS-CoV-2 infection is independently associated to increased protection against omicron, largely mediated by mucosal spike specific IgA responses (nested models lr test p = 0.02 and 0.008). Only 10% of infected participants remain asymptomatic through the course of their infection. We demonstrate that high levels of vaccine-induced spike-specific WT antibodies are linked to increased protection against infection and to reduced viral load if infected, and suggest that the additional protection offered by pre-omicron SARS-CoV-2 infection largely is mediated by mucosal spike-specific IgA.
Collapse
Affiliation(s)
- Ulrika Marking
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Sebastian Havervall
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Nina Greilert Norin
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Oscar Bladh
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Wanda Christ
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Max Gordon
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Henry Ng
- Department of Medical Cell Biology and SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Kim Blom
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Mia Phillipson
- Department of Medical Cell Biology and SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Sara Mangsbo
- Department of Pharmacy and SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Jessica J Alm
- Department of Microbiology, Tumor and Cell Biology & National Pandemic Center, Karolinska Institutet, Solna, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Nilsson
- Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Sophia Hober
- Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry and SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Jonas Klingström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Charlotte Thålin
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden.
| |
Collapse
|
35
|
Strathdee SA, Abramovitz D, Vera CF, Artamonova I, Patterson TL, Smith DM, Chaillon A, Bazzi AR. Predictors of COVID-19 vaccine uptake among people who inject drugs. Vaccine 2023; 41:1916-1924. [PMID: 36697311 PMCID: PMC9868393 DOI: 10.1016/j.vaccine.2023.01.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 12/21/2022] [Accepted: 01/18/2023] [Indexed: 01/24/2023]
Abstract
INTRODUCTION We studied characteristics of COVID-19 vaccination uptake among people who inject drugs (PWID). METHODS Participants aged ≥18 years who injected drugs ≤1 month ago were recruited into a community-based cohort from October 2020 to September 2021 in San Diego, California Poisson regression identified correlates of having had ≥1 COVID-19 vaccine dose based on semi-annual follow-up interviews through March 15, 2022. RESULTS Of 360 participants, 74.7% were male, mean age was 42 years; 63.1% were Hispanic/Mexican/Latinx. More than one-third had ≥1 co-morbidity. HIV and HCV seroprevalence were 4.2% and 50.6% respectively; 41.1% lacked health insurance. Only 37.8% reported having ≥1 COVID-19 vaccine dose. None received ≥3 doses. However, of those vaccinated, 37.5% were previously unwilling/unsure about COVID-19 vaccines. Believing COVID-19 vaccines include tracking devices (adjusted incidence rate ratio [aIRR]: 0.62; 95% CI: 0.42,0.92) and lacking health insurance (aIRR: 0.60; 95% CI: 0.40,0.91) were associated with approximately 40% lower COVID-19 vaccination rates). Ever receiving influenza vaccines (aIRR: 2.16; 95%CI: 1.46, 3.20) and testing HIV-seropositive (aIRR: 2.51; 95% CI: 1.03, 6.10) or SARS-CoV-2 RNA-positive (aIRR: 1.82; 95% CI: 1.05, 3.16) independently predicted higher COVID-19 vaccination rates. Older age, knowing more vaccinated people, and recent incarceration were also independently associated with higher COVID-19 vaccination rates. CONCLUSIONS One year after COVID-19 vaccines became available to U.S. adults, only one third of PWID had received ≥1 COVID-19 vaccine dose. Multi-faceted approaches that dispel disinformation, integrate public health and social services and increase access to free, community-based COVID-19 vaccines are urgently needed.
Collapse
Affiliation(s)
- Steffanie A Strathdee
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Daniela Abramovitz
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Carlos F Vera
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Irina Artamonova
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Thomas L Patterson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
| | - Davey M Smith
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Antoine Chaillon
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Angela R Bazzi
- Herbert Wertheim School of Public Health, University of California San Diego, La Jolla, CA, USA; Department of Community Health Sciences, Boston University School of Public Health, Boston, MA, USA.
| |
Collapse
|
36
|
Abstract
SARS-CoV-2 viral load and detection of infectious virus in the respiratory tract are the two key parameters for estimating infectiousness. As shedding of infectious virus is required for onward transmission, understanding shedding characteristics is relevant for public health interventions. Viral shedding is influenced by biological characteristics of the virus, host factors and pre-existing immunity (previous infection or vaccination) of the infected individual. Although the process of human-to-human transmission is multifactorial, viral load substantially contributed to human-to-human transmission, with higher viral load posing a greater risk for onward transmission. Emerging SARS-CoV-2 variants of concern have further complicated the picture of virus shedding. As underlying immunity in the population through previous infection, vaccination or a combination of both has rapidly increased on a global scale after almost 3 years of the pandemic, viral shedding patterns have become more distinct from those of ancestral SARS-CoV-2. Understanding the factors and mechanisms that influence infectious virus shedding and the period during which individuals infected with SARS-CoV-2 are contagious is crucial to guide public health measures and limit transmission. Furthermore, diagnostic tools to demonstrate the presence of infectious virus from routine diagnostic specimens are needed.
Collapse
Affiliation(s)
- Olha Puhach
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Benjamin Meyer
- Centre for Vaccinology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Isabella Eckerle
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Geneva Centre for Emerging Viral Diseases, Geneva University Hospitals, Geneva, Switzerland.
- Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland.
| |
Collapse
|
37
|
Lee E, Park S, Choi JP, Kim MK, Yang E, Ham SY, Lee S, Lee B, Yang JS, Park BK, Kim DS, Lee SY, Lee JY, Jang HC, Jeon J, Park SW. Short-Term Effectiveness of Oral Nirmatrelvir/Ritonavir Against the SARS-CoV-2 Omicron Variant and Culture-Positive Viral Shedding. J Korean Med Sci 2023; 38:e59. [PMID: 36852855 PMCID: PMC9970787 DOI: 10.3346/jkms.2023.38.e59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/22/2022] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Information on the effectiveness of nirmatrelvir/ritonavir against the omicron is limited. The clinical response and viral kinetics to therapy in the real world need to be evaluated. METHODS Mild to moderate coronavirus disease 2019 (COVID-19) patients with risk factors for severe illness were prospectively enrolled as a treatment group with nirmatrelvir/ritonavir therapy versus a control group with supportive care. Serial viral load and culture from the upper respiratory tract were evaluated for seven days, and clinical responses and adverse reactions were evaluated for 28 days. RESULTS A total of 51 patients were analyzed including 40 in the treatment group and 11 in the control group. Faster symptom resolution during hospitalization (P = 0.048) was observed in the treatment group. Only minor adverse reactions were reported in 27.5% of patients. The viral load on Day 7 was lower in the treatment group (P = 0.002). The viral culture showed a positivity of 67.6% (25/37) vs. 100% (6/6) on Day 1, 0% (0/37) vs. 16.7 (1/6) on Day 5, and 0% (0/16) vs. 50.0% (2/4) on Day 7 in the treatment and control groups, respectively. CONCLUSIONS Nirmatrelvir/ritonavir against the omicron was safe and resulted in negative viral culture conversion after Day 5 of treatment with better symptomatic resolution.
Collapse
Affiliation(s)
- Eunyoung Lee
- Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Sehee Park
- National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Jae-Phil Choi
- Division of Infectious Diseases, Seoul Medical Center, Seoul, Korea
| | - Min-Kyung Kim
- Division of Infectious Diseases, National Medical Center, Seoul, Korea
| | - Eunmi Yang
- Division of Infectious Diseases, Seoul Medical Center, Seoul, Korea
| | - Sin Young Ham
- Seoul Veterans Hospital Medical Center, Seoul, Korea
| | - Seungjae Lee
- Seoul Veterans Hospital Medical Center, Seoul, Korea
| | - Bora Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Medical Center, Seoul, Korea
| | - Jeong-Sun Yang
- National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Byoung Kwon Park
- National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Da Sol Kim
- National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - So-Young Lee
- National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Joo-Yeon Lee
- National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Hee-Chang Jang
- National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Jaehyun Jeon
- Division of Infectious Diseases, National Medical Center, Seoul, Korea.
| | - Sang-Won Park
- Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea.
| |
Collapse
|
38
|
Yahata H, Kato K, Shimokawa M, Kawamura K, Shimono N, Kawana K, Okamoto A, Aoki D, Kimura T. Study of the effects of in-person attendance at academic conferences on the health of the attendees under COVID-19 pandemic. J Obstet Gynaecol Res 2023; 49:1083-1089. [PMID: 36812688 DOI: 10.1111/jog.15626] [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: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023]
Abstract
OBJECTIVE To analyze the effects of in-person attendance at an academic conference held during the Covid-19 pandemic on the health of the attendees, as assessed based on symptoms such as fever and cough attributed to infection with the Covid-19 virus. METHODS A questionnaire was used to survey the members of the Japan Society of Obstetrics and Gynecology (JSOG) about their health during the period from August 7 to August 12, 2022, after the 74th Annual Congress of the JSOG, which was held August 5 to 7. RESULTS Our survey yielded responses from 3054 members (1566 of whom had attended the congress in person and 1488 of whom had not attended in person); 102 (6.5%) of the in-person attendees and 93 (6.2%) of the people who did not attend in person reported problems with their health. No statistically significant difference was found between these two groups (p = 0.766). In a univariate analysis of factors affecting the presence of health problems, attendees with age ≥60 years had significantly fewer health problems than attendees who were in their 20s (odds ratio: 0.366 [0.167-0.802; p = 0.0120]). In a multivariate analysis, attendees who had received four vaccine shots had significantly fewer health problems than attendees who had received three shots (odds ratio: 0.397 [0.229-0.690, p = 0.0010]). CONCLUSION Congress attendees who took precautions at the congress to avoid being infected and who had a high vaccination rate did not develop significantly more health problems associated with in-person attendance at the congress.
Collapse
Affiliation(s)
- Hideaki Yahata
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Kiyoko Kato
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Keiko Kawamura
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Nobuyuki Shimono
- Center for the Study of Global Infection, Kyushu University Hospital, Fukuoka, Japan
| | - Kei Kawana
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Tokyo, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, Jikei University School of Medicine, Tokyo, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
39
|
Milito C, Firinu D, Bez P, Villa A, Punziano A, Lagnese G, Costanzo G, van Leeuwen LPM, Piazza B, Deiana CM, d’Ippolito G, Del Giacco SR, Rattazzi M, Spadaro G, Quinti I, Scarpa R, Dalm VASH, Cinetto F. A beacon in the dark: COVID-19 course in CVID patients from two European countries: Different approaches, similar outcomes. Front Immunol 2023; 14:1093385. [PMID: 36845159 PMCID: PMC9944020 DOI: 10.3389/fimmu.2023.1093385] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/09/2023] [Indexed: 02/10/2023] Open
Abstract
Background CVID patients present an increased risk of prolonged SARS-CoV-2 infection and re-infection and a higher COVID-19-related morbidity and mortality compared to the general population. Since 2021, different therapeutic and prophylactic strategies have been employed in vulnerable groups (vaccination, SARS-CoV-2 monoclonal antibodies and antivirals). The impact of treatments over the last 2 years has not been explored in international studies considering the emergence of viral variants and different management between countries. Methods A multicenter retrospective/prospective real-life study comparing the prevalence and outcomes of SARS-CoV-2 infection between a CVID cohort from four Italian Centers (IT-C) and one cohort from the Netherlands (NL-C), recruiting 773 patients. Results 329 of 773 CVID patients were found positive for SARS-CoV-2 infection between March 1st, 2020 and September 1st 2022. The proportion of CVID patients infected was comparable in both national sub-cohorts. During all waves, chronic lung disease, "complicated" phenotype, chronic immunosuppressive treatment and cardiovascular comorbidities impacted on hospitalization, whereas risk factors for mortality were older age, chronic lung disease, and bacterial superinfections. IT-C patients were significantly more often treated, both with antivirals and mAbs, than NL-C patients. Outpatient treatment, available only in Italy, started from the Delta wave. Despite this, no significant difference was found for COVID-19 severity between the two cohorts. However, pooling together specific SARS-CoV-2 outpatient treatments (mAbs and antivirals), we found a significant effect on the risk of hospitalization starting from Delta wave. Vaccination with ≥ 3 doses shortened RT-PCR positivity, with an additional effect only in patients receiving antivirals. Conclusions The two sub-cohorts had similar COVID-19 outcomes despite different treatment approaches. This points out that specific treatment should now be reserved for selected subgroups of CVID patients, based on pre-existing conditions.
Collapse
Affiliation(s)
- Cinzia Milito
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Davide Firinu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Patrick Bez
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Annalisa Villa
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandra Punziano
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Gianluca Lagnese
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Giulia Costanzo
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Leanne P. M. van Leeuwen
- Department of Viroscience, Travel Clinic, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Beatrice Piazza
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Carla Maria Deiana
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | | | - Marcello Rattazzi
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Riccardo Scarpa
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy,*Correspondence: Riccardo Scarpa,
| | - Virgil A. S. H. Dalm
- Department of Internal Medicine, Division of Allergy and Clinical Immunology, Department of Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Francesco Cinetto
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy
| |
Collapse
|
40
|
Development and validation of a nomogram to predict failure of 14-day negative nucleic acid conversion in adults with non-severe COVID-19 during the Omicron surge: a retrospective multicenter study. Infect Dis Poverty 2023; 12:7. [PMID: 36750862 PMCID: PMC9902821 DOI: 10.1186/s40249-023-01057-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/17/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND With the variability in emerging data, guidance on the isolation duration for patients with coronavirus disease 2019 (COVID-19) due to the Omicron variant is controversial. This study aimed to determine the predictors of prolonged viral RNA shedding in patients with non-severe COVID-19 and construct a nomogram to predict patients at risk of 14-day PCR conversion failure. METHODS Adult patients with non-severe COVID-19 were enrolled from three hospitals of eastern China in Spring 2022. Viral shedding time (VST) was defined as either the day of the first positive test or the day of symptom onset, whichever was earlier, to the date of the first of two consecutively negative PCR tests. Patients from one hospital (Cohort I, n = 2033) were randomly grouped into training and internal validation sets. Predictors of 14-day PCR conversion failure were identified and a nomogram was developed by multivariable logistic regression using the training dataset. Two hospitals (Cohort II, n = 1596) were used as an external validation set to measure the performance of this nomogram. RESULTS Of the 2033 patients from Cohort I, the median VST was 13.0 (interquartile range: 10.0‒16.0) days; 716 (35.2%) lasted > 14 days. In the training set, increased age [per 10 years, odds ratio (OR) = 1.29, 95% confidence interval (CI): 1.15‒1.45, P < 0.001] and high Charlson comorbidity index (OR = 1.25, 95% CI: 1.08‒1.46, P = 0.004) were independent risk factors for VST > 14 days, whereas full or boosted vaccination (OR = 0.63, 95% CI: 0.42‒0.95, P = 0.028) and antiviral therapy (OR = 0.56, 95% CI: 0.31‒0.96, P = 0.040) were protective factors. These predictors were used to develop a nomogram to predict VST > 14 days, with an area under the ROC curve (AUC) of 0.73 in the training set (AUC, 0.74 in internal validation set; 0.76 in external validation set). CONCLUSIONS Older age, increasing comorbidities, incomplete vaccinations, and lack of antiviral therapy are risk factors for persistent infection with Omicron variant for > 14 days. A nomogram based on these predictors could be used as a prediction tool to guide treatment and isolation strategies.
Collapse
|
41
|
Nielsen SS, Alvarez J, Bicout DJ, Calistri P, Canali E, Drewe JA, Garin‐Bastuji B, Gonzales Rojas JL, Gortázar C, Herskin M, Michel V, Miranda Chueca MÁ, Padalino B, Pasquali P, Roberts HC, Spoolder H, Velarde A, Viltrop A, Winckler C, Adlhoch C, Aznar I, Baldinelli F, Boklund A, Broglia A, Gerhards N, Mur L, Nannapaneni P, Ståhl K. SARS-CoV-2 in animals: susceptibility of animal species, risk for animal and public health, monitoring, prevention and control. EFSA J 2023; 21:e07822. [PMID: 36860662 PMCID: PMC9968901 DOI: 10.2903/j.efsa.2023.7822] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
The epidemiological situation of SARS-CoV-2 in humans and animals is continually evolving. To date, animal species known to transmit SARS-CoV-2 are American mink, raccoon dog, cat, ferret, hamster, house mouse, Egyptian fruit bat, deer mouse and white-tailed deer. Among farmed animals, American mink have the highest likelihood to become infected from humans or animals and further transmit SARS-CoV-2. In the EU, 44 outbreaks were reported in 2021 in mink farms in seven MSs, while only six in 2022 in two MSs, thus representing a decreasing trend. The introduction of SARS-CoV-2 into mink farms is usually via infected humans; this can be controlled by systematically testing people entering farms and adequate biosecurity. The current most appropriate monitoring approach for mink is the outbreak confirmation based on suspicion, testing dead or clinically sick animals in case of increased mortality or positive farm personnel and the genomic surveillance of virus variants. The genomic analysis of SARS-CoV-2 showed mink-specific clusters with a potential to spill back into the human population. Among companion animals, cats, ferrets and hamsters are those at highest risk of SARS-CoV-2 infection, which most likely originates from an infected human, and which has no or very low impact on virus circulation in the human population. Among wild animals (including zoo animals), mostly carnivores, great apes and white-tailed deer have been reported to be naturally infected by SARS-CoV-2. In the EU, no cases of infected wildlife have been reported so far. Proper disposal of human waste is advised to reduce the risks of spill-over of SARS-CoV-2 to wildlife. Furthermore, contact with wildlife, especially if sick or dead, should be minimised. No specific monitoring for wildlife is recommended apart from testing hunter-harvested animals with clinical signs or found-dead. Bats should be monitored as a natural host of many coronaviruses.
Collapse
|
42
|
Tan ST, Kwan AT, Rodríguez-Barraquer I, Singer BJ, Park HJ, Lewnard JA, Sears D, Lo NC. Infectiousness of SARS-CoV-2 breakthrough infections and reinfections during the Omicron wave. Nat Med 2023; 29:358-365. [PMID: 36593393 PMCID: PMC9974584 DOI: 10.1038/s41591-022-02138-x] [Citation(s) in RCA: 111] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/18/2022] [Indexed: 01/03/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) breakthrough infections in vaccinated individuals and reinfections in previously infected individuals have become increasingly common. Such infections highlight a broader need to understand the contribution of vaccination, including booster doses, and natural immunity to the infectiousness of individuals with SARS-CoV-2 infections, especially in high-risk populations with intense transmission, such as in prisons. Here we show that both vaccine-derived and naturally acquired immunity independently reduce the infectiousness of persons with Omicron variant SARS-CoV-2 infections in a prison setting. Analyzing SARS-CoV-2 surveillance data from December 2021 to May 2022 across 35 California state prisons with a predominately male population, we estimate that unvaccinated Omicron cases had a 36% (95% confidence interval (CI): 31-42%) risk of transmitting infection to close contacts, as compared to a 28% (25-31%) risk among vaccinated cases. In adjusted analyses, we estimated that any vaccination, prior infection alone and both vaccination and prior infection reduced an index case's risk of transmitting infection by 22% (6-36%), 23% (3-39%) and 40% (20-55%), respectively. Receipt of booster doses and more recent vaccination further reduced infectiousness among vaccinated cases. These findings suggest that, although vaccinated and/or previously infected individuals remain highly infectious upon SARS-CoV-2 infection in this prison setting, their infectiousness is reduced compared to individuals without any history of vaccination or infection. This study underscores benefit of vaccination to reduce, but not eliminate, transmission.
Collapse
Affiliation(s)
- Sophia T Tan
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Ada T Kwan
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, CA, USA.,Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Isabel Rodríguez-Barraquer
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA.,Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Benjamin J Singer
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Hailey J Park
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Joseph A Lewnard
- Division of Epidemiology and Biostatistics, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.,Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.,Center for Computational Biology, College of Engineering, University of California, Berkeley, Berkeley, CA, USA
| | - David Sears
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.,Division of Infectious Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Nathan C Lo
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA. .,Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
43
|
Doke PP, Mhaske ST, Oka G, Kulkarni R, Muley V, Mishra AC, Arankalle VA. SARS-CoV-2 breakthrough infections during the second wave of COVID-19 at Pune, India. Front Public Health 2023; 10:1040012. [PMID: 36711329 PMCID: PMC9877521 DOI: 10.3389/fpubh.2022.1040012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
Breakthrough infections following SARS-CoV-2 vaccination remain the global concern. The current study was conducted during the second wave of COVID-19 (1st March-7th July 2021) in Pune, India, at two tertiary care hospitals. Of the 6,159 patients diagnosed as COVID-19, 372/2,210 (16.8%) were breakthrough infections. Of these, 81.1 and 18.8% received one or two doses of Covishield or Covaxin, respectively. Of note, 30.7% patients were with comorbidities, hypertension being the commonest (12.44%). The majority of infections were mild (81.2%). Forty-three patients with breakthrough infections were hospitalized with severe (n = 27, 62.8%) or moderate (n = 16, 37.2%) disease. The receptor binding domain (RBD) sequences from vaccinated (n = 126) and non-vaccinated (n = 168) samples were used for variant analysis. The delta variant was predominant followed by kappa in both vaccinated and non-vaccinated groups. Viral load (qRT-PCR) was not different among these categories. Full-genome comparisons of sequences in relation to vaccination status did not identify any mutation characteristic of the vaccinated group. Irrespective of the number of doses, neutralizing antibody titers (PRNT50) during the first week of clinical disease were higher in the vaccinated patients than the unvaccinated category. In conclusion, though not completely, SARS-CoV-2 vaccines used for country-wide immunization did reduce disease severity among the individuals without any comorbidity by inducing rapid immune response against distinctly different delta and kappa variants. The utility against emerging variants with further mutations need to be carefully examined.
Collapse
Affiliation(s)
- Prakash P. Doke
- Department of Community Medicine, Bharati Vidyapeeth (Deemed to be University) Medical College, Pune, India
| | - Suhas T. Mhaske
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Gauri Oka
- Department of Community Medicine, Bharati Vidyapeeth (Deemed to be University) Medical College, Pune, India
| | - Ruta Kulkarni
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Vrishali Muley
- Department of Microbiology, Smt. Kashibai Navale Medical College, Pune, India
| | - Akhilesh Chandra Mishra
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Vidya A. Arankalle
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| |
Collapse
|
44
|
Dhakal S, Yu T, Yin A, Pisanic N, Demko ZO, Antar AAR, Cox AL, Heaney CD, Manabe YC, Klein SL. Reconsideration of Antinucleocapsid IgG Antibody as a Marker of SARS-CoV-2 Infection Postvaccination for Mild COVID-19 Patients. Open Forum Infect Dis 2023; 10:ofac677. [PMID: 36655185 PMCID: PMC9835753 DOI: 10.1093/ofid/ofac677] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Antinucleocapsid (anti-N) immunoglobulin G antibody responses were lower in plasma and oral fluid after severe acute respiratory syndrome coronavirus 2 infection in vaccinated patients compared with patients infected before vaccination or infected without vaccination. This raises questions about the long-term use of anti-N antibodies as a marker for natural infection for surveillance.
Collapse
Affiliation(s)
- Santosh Dhakal
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Tong Yu
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Anna Yin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Nora Pisanic
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Zoe O Demko
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Annukka A R Antar
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Andrea L Cox
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Christopher D Heaney
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Yukari C Manabe
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
45
|
Kang S, Kim JY, Park H, Lim SY, Kim J, Chang E, Bae S, Jung J, Kim MJ, Chong YP, Lee S, Choi S, Kim YS, Park M, Kim S. Comparison of secondary attack rate and viable virus shedding between patients with SARS-CoV-2 Delta and Omicron variants: A prospective cohort study. J Med Virol 2023; 95:e28369. [PMID: 36458559 PMCID: PMC9877691 DOI: 10.1002/jmv.28369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/12/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022]
Abstract
There are limited data comparing the transmission rates and kinetics of viable virus shedding of the Omicron variant to those of the Delta variant. We compared these rates in hospitalized patients infected with Delta and Omicron variants. We prospectively enrolled adult patients with COVID-19 admitted to a tertiary care hospital in South Korea between September 2021 and May 2022. Secondary attack rates were calculated by epidemiologic investigation, and daily saliva samples were collected to evaluate viral shedding kinetics. Genomic and subgenomic SARS-CoV-2 RNA was measured by PCR, and virus culture was performed from daily saliva samples. A total of 88 patients with COVID-19 who agreed to daily sampling and were interviewed, were included. Of the 88 patients, 48 (59%) were infected with Delta, and 34 (41%) with Omicron; a further 5 patients gave undetectable or inconclusive RNA PCR results and 1 was suspected of being coinfected with both variants. Omicron group had a higher secondary attack rate (31% [38/124] vs. 7% [34/456], p < 0.001). Survival analysis revealed that shorter viable virus shedding period was observed in Omicron variant compared with Delta variant (median 4, IQR [1-7], vs. 8.5 days, IQR [5-12 days], p < 0.001). Multivariable analysis revealed that moderate-to-critical disease severity (HR: 1.96), and immunocompromised status (HR: 2.17) were independent predictors of prolonged viral shedding, whereas completion of initial vaccine series or first booster-vaccinated status (HR: 0.49), and Omicron infection (HR: 0.44) were independently associated with shorter viable virus shedding. Patients with Omicron infections had higher transmission rates but shorter periods of transmissible virus shedding than those with Delta infections.
Collapse
Affiliation(s)
- Sung‐Woon Kang
- Department of Infectious Diseases, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Ji Yeun Kim
- Department of Infectious Diseases, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Heedo Park
- Department of Biomedical Sciences, BK21 Graduate ProgramKorea University College of MedicineSeoulRepublic of Korea
- Department of Microbiology, Institute for Viral Diseases, Vaccine Innovation Center, College of MedicineKorea UniversitySeoulSouth Korea
| | - So Yun Lim
- Department of Infectious Diseases, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Jeonghun Kim
- Department of Biomedical Sciences, BK21 Graduate ProgramKorea University College of MedicineSeoulRepublic of Korea
- Department of Microbiology, Institute for Viral Diseases, Vaccine Innovation Center, College of MedicineKorea UniversitySeoulSouth Korea
| | - Euijin Chang
- Department of Infectious Diseases, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Seongman Bae
- Department of Infectious Diseases, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Jiwon Jung
- Department of Infectious Diseases, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Min Jae Kim
- Department of Infectious Diseases, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Yong Pil Chong
- Department of Infectious Diseases, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Sang‐Oh Lee
- Department of Infectious Diseases, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Sang‐Ho Choi
- Department of Infectious Diseases, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Yang Soo Kim
- Department of Infectious Diseases, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Man‐Seong Park
- Department of Biomedical Sciences, BK21 Graduate ProgramKorea University College of MedicineSeoulRepublic of Korea
- Department of Microbiology, Institute for Viral Diseases, Vaccine Innovation Center, College of MedicineKorea UniversitySeoulSouth Korea
| | - Sung‐Han Kim
- Department of Infectious Diseases, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| |
Collapse
|
46
|
Tsang NNY, So HC, Cowling BJ, Leung GM, Ip DKM. Effectiveness of BNT162b2 and CoronaVac COVID-19 vaccination against asymptomatic and symptomatic infection of SARS-CoV-2 omicron BA.2 in Hong Kong: a prospective cohort study. THE LANCET. INFECTIOUS DISEASES 2022; 23:421-434. [PMID: 36521506 PMCID: PMC9744442 DOI: 10.1016/s1473-3099(22)00732-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/18/2022] [Accepted: 10/27/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND COVID-19 vaccines provide protection against symptomatic infection that might require medical attention and against severe outcomes; however, there is a paucity of evidence regarding the effectiveness of the BNT162b2 and CoronaVac vaccines and their booster regimens against asymptomatic or mild omicron infections in the community. We aimed to measure the effectiveness of BNT162b2 and CoronaVac vaccines against asymptomatic and symptomatic SARS-CoV-2 omicron infections, during a period of omicron BA.2 predominance in Hong Kong. METHODS In this prospective cohort study in a population that was generally infection-naive before the large omicron BA.2 wave between January and late May, 2022, we established a public health surveillance platform to monitor the evolving activity of SARS-CoV-2 infections in the community. We recruited a cohort of individuals aged 5 years and older between March 1 and March 7, 2022, from the general population. Individuals were enrolled from all 18 districts of Hong Kong, according to a predefined age-stratified quota, primarily by random digit dialing (generating suitable eight-digit local telephone numbers by randomly picking sets of the first four digits from a sampling frame, and randomly generating the last four digits), and supplemented by our existing cohorts (which included cohorts for studying influenza vaccination from school-based vaccination programmes and cohorts for SARS-CoV-2 seroprevalence from the community), to ensure representativeness of the population in Hong Kong. Participants did weekly rapid antigen testing with a self-collected pooled nasal and throat swab, regardless of symptom and exposure status, from March 1 to April 15, 2022. Individuals reporting a history of SARS-CoV-2 infection confirmed by laboratory PCR testing before enrolment were excluded from the vaccine effectiveness analysis to avoid potential bias due to infection-induced immunity. The primary outcomes of the study were the incidence of SARS-CoV-2 infection, including asymptomatic and symptomatic infections, and the vaccine effectiveness of BNT162b2 and CoronaVac vaccines. The effectiveness of one, two, and three doses of vaccination was estimated with a Cox proportional hazards regression model with time-dependent covariates, allowing for changes in vaccination status over time, after adjustment for demographic factors and pre-existing medical conditions. FINDINGS Of the 8636 individuals included in the analysis, 7233 (84%) received at least two doses of vaccine, 3993 (46%) received booster doses, and 903 (10%) reported SARS-CoV-2 infection. Among these infections 589 (65·2%) were symptomatic and 314 (34·8%) were asymptomatic at the time of testing. Statistically significant protection against asymptomatic and symptomatic SARS-CoV-2 omicron infection was found only for those who received a BNT162b2 or CoronaVac booster dose, with a vaccine effectiveness of 41·4% (23·2 to 55·2; p=0·0001) and 32·4% (9·0 to 49·8; p=0·0098), respectively. The vaccine effectiveness of BNT162b2 and CoronaVac boosters was further increased to 50·9% (95% CI 31·0-65·0; p<0·0001) and 41·6% (15·0-59·8; p=0·0049), respectively, for symptomatic omicron infections. A similar pattern of vaccine effectiveness (55·8%, 22·9-74·6; p=0·0040) was also conferred after receipt of a BNT162b2 booster by individuals who received a CoronaVac primary vaccination series. INTERPRETATION Two doses of either vaccine did not provide significant protection against COVID-19 infection. However, receipt of a BNT162b2 booster or CoronaVac booster was associated with a significantly lower risk of omicron BA.2 infection and symptomatic infection. Our findings confirm the effectiveness of booster doses to protect against mild and asymptomatic infection. FUNDING Henry Fok Foundation and Hong Kong Health Bureau.
Collapse
Affiliation(s)
- Nicole Ngai Yung Tsang
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Hau Chi So
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Benjamin J Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Gabriel M Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Dennis Kai Ming Ip
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
47
|
Clinical scoring system to predict viable viral shedding in patients with COVID-19. J Clin Virol 2022; 157:105319. [PMID: 36223658 PMCID: PMC9529675 DOI: 10.1016/j.jcv.2022.105319] [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: 06/30/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND The Centers for Disease Control and Prevention (CDC) recommends 5-10 days of isolation for patients with COVID-19, depending on symptom duration and severity. However, in clinical practice, an individualized approach is required. We thus developed a clinical scoring system to predict viable viral shedding. METHODS We prospectively enrolled adult patients with SARS-CoV-2 infection admitted to a hospital or community isolation facility between February 2020 and January 2022. Daily dense respiratory samples were obtained, and genomic RNA viral load assessment and viral culture were performed. Clinical predictors of negative viral culture results were identified using survival analysis and multivariable analysis. RESULTS Among 612 samples from 121 patients including 11 immunocompromised patients (5 organ transplant recipients, 5 with hematologic malignancy, and 1 receiving immunosuppressive agents) with varying severity, 154 (25%) revealed positive viral culture results. Multivariable analysis identified symptom onset day, viral copy number, disease severity, organ transplant recipient, and vaccination status as independent predictors of culture-negative rate. We developed a 4-factor predictive model based on viral copy number (-3 to 3 points), disease severity (1 point for moderate to critical disease), organ transplant recipient (2 points), and vaccination status (-2 points for fully vaccinated). Predicted culture-negative rates were calculated through the symptom onset day and the score of the day the sample was collected. CONCLUSIONS Our clinical scoring system can provide the objective probability of a culture-negative state in a patient with COVID-19 and is potentially useful for implementing personalized de-isolation policies beyond the simple symptom-based isolation strategy.
Collapse
|
48
|
Kandel C, Lee Y, Taylor M, Llanes A, McCready J, Crowl G, Powis J, Li AX, Shigayeva A, Yip L, Katz K, Kozak R, Mubareka S, McGeer A. Viral dynamics of the SARS-CoV-2 Omicron Variant among household contacts with 2 or 3 COVID-19 vaccine doses. J Infect 2022; 85:666-670. [PMID: 36283495 PMCID: PMC9595488 DOI: 10.1016/j.jinf.2022.10.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVES SARS-CoV-2 shedding has changed as new variants have emerged. It is important to understand the trajectory of PCR positivity due to Omicron in vaccinated populations. METHODS Double- or triple-vaccinated adult household contacts of individuals with COVID-19 self-collected oral-nasal swabs for 14 days. A hierarchical linear model estimated viral load trajectories and an exploratory logistic regression model assessed for factors associated with viral detection before symptom onset. RESULTS Forty-one participants developed COVID-19 with 37 (90%) symptomatic. Viral load peaked 3 days after symptom onset at a median concentration of 8.83 log10 copies/milliliter (range 5.95-10.32) and the mean difference between participants with two or three COVID-19 vaccine doses was 0.02 log10 copies/milliliter (95% CI -0.13 to 0.16). PCR positivity began with a range of 4 days prior to 3 days after symptom onset and was positive on the day of symptom onset in 76% (28/37). SARS-CoV-2 detection on the day of symptom onset was less likely among those with 2 vaccine doses (OR 0.13, 95%CI 0.02-0.79). 68% (25/37) of infected participants had detectable SARS-CoV-2 with Ct<30 at 7 days after symptom onset. CONCLUSIONS Peak viral load and duration of PCR positivity were similar in participants with COVID-19 after two versus three COVID-19 vaccine doses. Onset of viral detection relative to symptom onset was variable.
Collapse
Affiliation(s)
- Christopher Kandel
- Toronto East Health Network, Michael Garron Hospital, 825 Coxwell Avenue, Toronto, Ontario M4C 3E7, Canada.
| | - Yaejin Lee
- Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, Ontario M4N 3M5, Canada,Department of Laboratory Medicine and Pathobiology, King's College Cir, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Maureen Taylor
- Toronto East Health Network, Michael Garron Hospital, 825 Coxwell Avenue, Toronto, Ontario M4C 3E7, Canada
| | - Andrea Llanes
- Toronto East Health Network, Michael Garron Hospital, 825 Coxwell Avenue, Toronto, Ontario M4C 3E7, Canada
| | - Janine McCready
- Toronto East Health Network, Michael Garron Hospital, 825 Coxwell Avenue, Toronto, Ontario M4C 3E7, Canada
| | - Gloria Crowl
- Toronto East Health Network, Michael Garron Hospital, 825 Coxwell Avenue, Toronto, Ontario M4C 3E7, Canada
| | - Jeff Powis
- Toronto East Health Network, Michael Garron Hospital, 825 Coxwell Avenue, Toronto, Ontario M4C 3E7, Canada
| | - Angel Xinliu Li
- Sinai Health System, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada
| | - Altynay Shigayeva
- Sinai Health System, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada
| | - Lily Yip
- Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, Ontario M4N 3M5, Canada
| | - Kevin Katz
- Department of Laboratory Medicine and Pathobiology, King's College Cir, University of Toronto, Toronto, Ontario M5S 1A8, Canada,North York General Hospital, 4001 Leslie St, North York, Ontario M2K 1E1, Canada,Shared Hospital Laboratory, 555 Finch Ave W, Toronto, Ontario M2R 1N5, Canada
| | - Robert Kozak
- Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, Ontario M4N 3M5, Canada,Department of Laboratory Medicine and Pathobiology, King's College Cir, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Samira Mubareka
- Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, Ontario M4N 3M5, Canada,Department of Laboratory Medicine and Pathobiology, King's College Cir, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Allison McGeer
- Department of Laboratory Medicine and Pathobiology, King's College Cir, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Sinai Health System, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada.
| |
Collapse
|
49
|
Dingemans J, van der Veer BMJW, Gorgels KMF, Hackert V, den Heijer CDJ, Hoebe CJPA, Savelkoul PHM, van Alphen LB. Investigating SARS-CoV-2 breakthrough infections per variant and vaccine type. Front Microbiol 2022; 13:1027271. [PMID: 36504818 PMCID: PMC9729533 DOI: 10.3389/fmicb.2022.1027271] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/02/2022] [Indexed: 11/27/2022] Open
Abstract
Breakthrough SARS-CoV-2 infections have been reported in fully vaccinated individuals, in spite of the high efficacy of the currently available vaccines, proven in trials and real-world studies. Several variants of concern (VOC) have been proffered to be associated with breakthrough infections following immunization. In this study, we investigated 378 breakthrough infections recorded between January and July 2021 and compared the distribution of SARS-CoV-2 genotypes identified in 225 fully vaccinated individuals to the frequency of circulating community lineages in the region of South Limburg (The Netherlands) in a week-by-week comparison. Although the proportion of breakthrough infections was relatively low and stable when the Alpha variant was predominant, the rapid emergence of the Delta variant lead to a strong increase in breakthrough infections, with a higher relative proportion of individuals vaccinated with Vaxzevria or Jcovden being infected compared to those immunized with mRNA-based vaccines. A significant difference in median age was observed when comparing fully vaccinated individuals with severe symptoms (83 years) to asymptomatic cases (46.5 years) or individuals with mild-to-moderate symptoms (42 years). There was no association between SARS-CoV-2 genotype or vaccine type and disease symptoms. Furthermore, the majority of adaptive mutations were concentrated in the N-terminal domain of the Spike protein, highlighting its role in immune evasion. Interestingly, symptomatic individuals harbored significantly higher SARS-CoV-2 loads than asymptomatic vaccinated individuals and breakthrough infections caused by the Delta variant were associated with increased viral loads compared to those caused by the Alpha variant. In addition, we investigated the role of the Omicron variant in causing breakthrough infections by analyzing 135 samples that were randomly selected for genomic surveillance during the transition period from Delta to Omicron. We found that the proportion of Omicron vs. Delta infections was significantly higher in individuals who received a booster vaccine compared to both unvaccinated and fully vaccinated individuals. Altogether, these results indicate that the emergence of the Delta variant and in particular Omicron has lowered the efficiency of particular vaccine types to prevent SARS-CoV-2 infections and that, although rare, the elderly are particularly at risk of becoming severely infected as the consequence of a breakthrough infection.
Collapse
Affiliation(s)
- Jozef Dingemans
- Department of Medical Microbiology, Infectious diseases and Infection prevention, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center (MUMC+), Maastricht, Netherlands,*Correspondence: Jozef Dingemans, ; Brian M. J. W. van der Veer,
| | - Brian M. J. W. van der Veer
- Department of Medical Microbiology, Infectious diseases and Infection prevention, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center (MUMC+), Maastricht, Netherlands,*Correspondence: Jozef Dingemans, ; Brian M. J. W. van der Veer,
| | - Koen M. F. Gorgels
- Department of Sexual Health, Infectious Diseases and Environmental Health, South Limburg Public Health Service, Heerlen, Netherlands
| | - Volker Hackert
- Department of Sexual Health, Infectious Diseases and Environmental Health, South Limburg Public Health Service, Heerlen, Netherlands,Department of Social Medicine, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Casper D. J. den Heijer
- Department of Sexual Health, Infectious Diseases and Environmental Health, South Limburg Public Health Service, Heerlen, Netherlands,Department of Social Medicine, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Christian J. P. A Hoebe
- Department of Medical Microbiology, Infectious diseases and Infection prevention, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center (MUMC+), Maastricht, Netherlands,Department of Sexual Health, Infectious Diseases and Environmental Health, South Limburg Public Health Service, Heerlen, Netherlands,Department of Social Medicine, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Paul H. M. Savelkoul
- Department of Medical Microbiology, Infectious diseases and Infection prevention, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center (MUMC+), Maastricht, Netherlands
| | - Lieke B. van Alphen
- Department of Medical Microbiology, Infectious diseases and Infection prevention, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center (MUMC+), Maastricht, Netherlands
| |
Collapse
|
50
|
Cheng CD, Zhao S, Jiang J, Lin N, Li P, Ning XH, Zhang S. Impact of the COVID-19 pandemic on cardiac implantable electronic device implantation in China: Insights from 2 years of changing pandemic conditions. Front Public Health 2022; 10:1031241. [PMID: 36483238 PMCID: PMC9723342 DOI: 10.3389/fpubh.2022.1031241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/04/2022] [Indexed: 11/23/2022] Open
Abstract
Background A substantial reduction in the number of cardiac implantable electronic device (CIED) implantation was reported in the early stages of the COVID-19 pandemic. None of the studies have yet explored changes in CIED implantation during the following pandemic. Objective To explore changes in CIED implantation during the COVID-19 pandemic from 2020 to 2021. Methods From 2019 to 2021, 177,263 patients undergone CIED implantation from 1,227 hospitals in China were included in the analysis. Generalized linear models measured the differences in CIED implantation in different periods. The relationship between changes in CIED implantation and COVID-19 cases was assessed by simple linear regression models. Results Compared with the pre-COVID-19 period, the monthly CIED implantation decreased by 17.67% (95% CI: 16.62-18.72%, p < 0.001) in 2020. In 2021, the monthly number of CIED implantation increased by 15.60% (95% CI: 14.34-16.85%, p < 0.001) compared with 2020. For every 10-fold increase in the number of COVID-19 cases, the monthly number of pacemaker implantation decreased by 429 in 2021, while it decreased by 676 in 2020. The proportion of CIED implantation in secondary medical centers increased from 52.84% in 2019 to 56.77% in 2021 (p < 0.001). For every 10-fold increase in regional accumulated COVID-19 cases, the proportion of CIED implantation in secondary centers increased by 6.43% (95% CI: 0.47-12.39%, p = 0.036). Conclusion The impact of the COVID-19 pandemic on the number of CIED implantation is diminishing in China. Improving the ability of secondary medical centers to undertake more operations may be a critical way to relieve the strain on healthcare resources during the epidemic.
Collapse
Affiliation(s)
- Chen-di Cheng
- State Key Laboratory of Cardiovascular Disease, Arrhythmia Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuang Zhao
- State Key Laboratory of Cardiovascular Disease, Arrhythmia Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang Jiang
- State Key Laboratory of Cardiovascular Disease, Arrhythmia Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Lin
- State Key Laboratory of Cardiovascular Disease, Arrhythmia Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Center for Cardiovascular Quality Improvement, Fuwai Hospital, National Center for Cardiovascular Diseases, Beijing, China
| | - Ping Li
- State Key Laboratory of Cardiovascular Disease, Arrhythmia Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Center for Cardiovascular Quality Improvement, Fuwai Hospital, National Center for Cardiovascular Diseases, Beijing, China
| | - Xiao-hui Ning
- State Key Laboratory of Cardiovascular Disease, Arrhythmia Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Center for Cardiovascular Quality Improvement, Fuwai Hospital, National Center for Cardiovascular Diseases, Beijing, China
| | - Shu Zhang
- State Key Laboratory of Cardiovascular Disease, Arrhythmia Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Center for Cardiovascular Quality Improvement, Fuwai Hospital, National Center for Cardiovascular Diseases, Beijing, China
| |
Collapse
|