1
|
Kakeya H, Nitta T, Kamijima Y, Miyazawa T. Significant Increase in Excess Deaths after Repeated COVID-19 Vaccination in Japan. JMA J 2025; 8:584-586. [PMID: 40416011 PMCID: PMC12095670 DOI: 10.31662/jmaj.2024-0298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/10/2025] [Indexed: 05/27/2025] Open
Abstract
Although Japan recorded the world's highest rate of COVID-19 messenger ribonucleic acid (mRNA) vaccination doses per capita, COVID-19 cases and deaths exploded after the emergence of the Omicron variant, followed by a significant increase in excess deaths in 2022 and 2023. Although several hypotheses have been proposed to explain these phenomena, the truth remains to be established because sufficient studies and data disclosures have not been conducted to adequately investigate the possible contribution of mRNA vaccines. The causes of the excess deaths from not only COVID-19 but also other factors after repeated mRNA vaccinations must be elucidated, given this could provide valuable information to help combat future infectious disease outbreaks.
Collapse
Affiliation(s)
- Hideki Kakeya
- Institute of Systems and Information Engineering, University of Tsukuba, Tsukuba, Japan
| | - Takeshi Nitta
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yukari Kamijima
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | | |
Collapse
|
2
|
Zahm AM, Cranney CW, Gormick AN, Rondem KE, Schmitz B, Himes SR, English JG. ConSeqUMI, an error-free nanopore sequencing pipeline to identify and extract individual nucleic acid molecules from heterogeneous samples. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.647077. [PMID: 40236236 PMCID: PMC11996460 DOI: 10.1101/2025.04.03.647077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Nanopore sequencing has revolutionized genetic analysis by offering linkage information across megabase-scale genomes. However, the high intrinsic error rate of nanopore sequencing impedes the analysis of complex heterogeneous samples, such as viruses, bacteria, complex libraries, and edited cell lines. Achieving high accuracy in single-molecule sequence identification would significantly advance the study of diverse genomic populations, where clonal isolation is traditionally employed for complete genomic frequency analysis. Here, we introduce ConSeqUMI, an innovative experimental and analytical pipeline designed to address long-read sequencing error rates using unique molecular indices for precise consensus sequence determination. ConSeqUMI processes nanopore sequencing data without the need for reference sequences, enabling accurate assembly of individual molecular sequences from complex mixtures. We establish robust benchmarking criteria for this platform's performance and demonstrate its utility across diverse experimental contexts, including mixed plasmid pools, recombinant adeno-associated virus genome integrity, and CRISPR/Cas9-induced genomic alterations. Furthermore, ConSeqUMI enables detailed profiling of human pathogenic infections, as shown by our analysis of SARS-CoV-2 spike protein variants, revealing substantial intra-patient genetic heterogeneity. Lastly, we demonstrate how individual clonal isolates can be extracted directly from sequencing libraries at low cost, allowing for post-sequencing identification and validation of observed variants. Our findings highlight the robustness of ConSeqUMI in processing sequencing data from UMI-labeled molecules, offering a critical tool for advancing genomic research. GRAPHICAL ABSTRACT
Collapse
|
3
|
Deming ME, Brown ER, McArthur MA, Schrag SJ, Arvay M, Humphrys M, Ravel J, Adelglass J, Essink B, Musante DB, Maguire R, Gorman R, Formentini E, Mason R, Robb ML, Neuzil KM, Rapaka RR, Wolff P, Kotloff KL. Vaccine efficacy of NVX-CoV2373 against SARS-CoV-2 infection in adolescents in the USA: an ancillary study to a phase 3, observer-blinded, randomised, placebo-controlled trial. THE LANCET. MICROBE 2025; 6:100984. [PMID: 39884302 DOI: 10.1016/j.lanmic.2024.100984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 02/01/2025]
Abstract
BACKGROUND Although existing COVID-19 vaccines are known to be highly effective against severe disease and death, data are needed to assess their ability to reduce SARS-CoV-2 infection. We aimed to estimate the efficacy of the NVX-CoV2373 protein subunit vaccine against SARS-CoV-2 infection, regardless of symptoms, among adolescents. METHODS We performed an ancillary observational study (SNIFF) to the phase 3, observer-blinded, randomised, placebo-controlled PREVENT-19 trial that assessed vaccine efficacy against symptomatic COVID-19 in the USA. Participants in the PREVENT-19 trial included healthy adolescents aged 12-17 years and with no history of laboratory-confirmed SARS-CoV-2 infection. They were randomly assigned (2:1) to receive either the NVX-CoV2373 (Novavax, Gaithersburg, MD, USA) vaccine (immediate NVX-CoV2373 group) or placebo (delayed NVX-CoV2373 group) on days 0 and 21 (initial series). After 2 months, in a crossover series, participants received two doses, 21 days apart, of the intervention that they did not receive in their initial series. Participants at 47 of the PREVENT-19 sites were invited to participate in the SNIFF study and self-collect nasal swabs at home twice weekly for SARS-CoV-2 testing to assess vaccine efficacy against SARS-CoV-2 infection. This primary outcome was defined as the first identification of SARS-CoV-2 detected by RT-PCR, regardless of symptoms, with onset within 4 weeks after the second dose of the initial vaccination series until the second dose of the crossover series. Secondary outcomes were vaccine efficacy against asymptomatic and minimally symptomatic SARS-CoV-2 infection, durability of vaccine efficacy against SARS-CoV-2 infection, and durability of vaccine efficacy against asymptomatic and minimally symptomatic infections. Outcomes were analysed in the modified intention-to-treat population, which included all participants without previous SARS-CoV-2 infection and was restricted to participants enrolled within 4 weeks of the second dose of the primary (primary analysis population) or crossover (post-crossover analysis population) series. This study is registered with ClinicalTrials.gov (NCT04611802). FINDINGS Between June 1 and Dec 17, 2021, 1196 (53·2%) of the 2247 adolescent participants recruited in the PREVENT-19 trial enrolled in the SNIFF study. The primary analysis population included 471 participants in the immediate NVX-CoV2373 group and 220 in the delayed NVX-CoV2373 group. Incidence of SARS-CoV-2 infection was 14·9 cases per 100 person-years (95% CI 7·9-25·5) in the immediate group and 54·2 cases per 100 person-years (33·6-82·9) in the delayed group; vaccine efficacy was 73·5% (95% CI 47·1-86·7; p=0·0002). Incidence of minimally symptomatic or asymptomatic SARS-CoV-2 infection was 10·3 cases per 100 person-years (95% CI 4·7-19·6) in the immediate group and 36·1 cases per 100 person-years (19·8-60·7) in the delayed group; vaccine efficacy was 72·8% (95% CI 37·1-88·2; p=0·0023). After the second crossover dose, incidence of SARS-CoV-2 was 14·6 cases per 100 person-years (95% CI 8·6-23·0) in the immediate group (receiving placebo at crossover) and 9·1 cases per 100 person-years (3·0-21·3) in the delayed group, with a durability ratio of 160·3 (95% CI 59·5-431·6; p=0·35). Almost all infections after crossover were minimally symptomatic or asymptomatic, with a durability ratio of 151·4 (55·9-410·4; p=0·41). INTERPRETATION Among adolescents participating in the PREVENT-19 trial during the delta (B.1.617.2) variant wave of the COVID-19 pandemic, the NVX-CoV2373 vaccine was highly efficacious against SARS-CoV-2 infection regardless of symptoms, indicating its potential to reduce the reservoir of infections that contribute to community transmission. FUNDING US Department of Health and Human Services, Administration for Strategic Preparedness and Response, Biomedical Advanced Research and Development Authority, National Institute of Allergy and Infectious Diseases, and National Institutes of Health.
Collapse
Affiliation(s)
- Meagan E Deming
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | - Monica A McArthur
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stephanie J Schrag
- US COVID-19 Domestic Response and Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Melissa Arvay
- US COVID-19 Domestic Response and Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Mike Humphrys
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | - Rebecca Maguire
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Richard Gorman
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | - Robin Mason
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Merlin L Robb
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rekha R Rapaka
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Peter Wolff
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Washington, DC, USA
| | - Karen L Kotloff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Effa EE, Ita O, Mwankon J, Siyanbade F, Iwomi F, Ochodo E, Villanueva G, Meremikwu MM. Post-exposure testing at healthcare facilities with SARS-CoV-2 transmission: A rapid review. J Public Health Afr 2025; 16:623. [PMID: 40083354 PMCID: PMC11905177 DOI: 10.4102/jphia.v16i2.623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 11/13/2024] [Indexed: 03/16/2025] Open
Abstract
Background Post-exposure severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) testing following health facility outbreaks may control the spread of infection. Aim This study aimed to assess the impact of testing for SARS-CoV-2 infection on health outcomes during healthcare facility outbreaks. Setting This review included studies conducted at skilled nursing facilities, a cancer centre, and a geriatric psychiatric facility. Methods We followed the methods for conducting rapid systematic reviews, searched databases from December 2019 to August 2022, assessed the risk of bias using the modified Newcastle Ottawa scale, and graded the certainty of evidence using the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) approach. We pooled the prevalence, mortality, and hospitalisation results as appropriate. Results Of the 3055 articles from database search, no study was eligible for inclusion as outlined in the protocol. However, eight non-comparative reports (case series) in skilled nursing facilities were included. The pooled prevalence of SARS-CoV-2 infection among residents of care homes and patients were 38% (95% confidence interval [CI] = 25% - 51%; 5 studies, 2044 participants; I 2 = 94%, very low certainty evidence) and was 12% (95% CI = 6% - 19%; 5 studies, 2312 participants; I 2 = 94%, very low certainty evidence) for exposed healthcare workers. The pooled mortality estimate and hospitalisation rate were 17% and 24%, respectively, (very low certainty evidence). Conclusion There is no identified evidence for or against testing of people in healthcare facilities where there is ongoing transmission of SARS-CoV-2 infection. Contribution The evaluation of the effectiveness of testing strategies during SARS-CoV-2 outbreaks need baseline and follow-up data from well-designed before and after studies appropriate for the setting.
Collapse
Affiliation(s)
- Emmanuel E Effa
- Department of Internal Medicine, Faculty of Clinical Sciences, University of Calabar, Calabar, Nigeria
| | - Okokon Ita
- Department of Medical Microbiology, Faculty of Laboratory Medicine, University of Calabar, Calabar, Nigeria
| | - Joshua Mwankon
- Department of Family Medicine, Faculty of Clinical Sciences, University of Calabar, Calabar, Nigeria
| | - Funmi Siyanbade
- School of Nursing, University of Calabar Teaching Hospital, Calabar, Nigeria
| | - Francis Iwomi
- Cochrane Nigeria, Calabar Institute of Tropical Disease Research and Prevention, University of Calabar Teaching Hospital, Calabar, Nigeria
| | - Eleanor Ochodo
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | | | - Martin M Meremikwu
- Department of Paediatrics, University of Calabar Teaching Hospital, Calabar, Nigeria
| |
Collapse
|
5
|
Robinson L, Feting A, Isozaki I, Seyfert-Morgolis V, Jay M, Kim E, Cairns C. Time-varying effects of COVID-19 vaccination on symptomatic and asymptomatic infections in a prospective university cohort in the USA. BMJ Open 2025; 15:e084408. [PMID: 39987006 PMCID: PMC11848656 DOI: 10.1136/bmjopen-2024-084408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/07/2025] [Indexed: 02/24/2025] Open
Abstract
OBJECTIVES Despite widespread vaccination programmes and consensus recommendations, the understanding of the durability of COVID-19 vaccination against ensuing infection and transmission at the individual level is incomplete. The objective of this study was to estimate the effects of time-varying covariates including time since vaccination and symptoms on subsequent positive SARS-CoV-2 test results and assess the stability of these effects between March 2020 and April 2022. DESIGN Prospective cohort study. SETTING Urban university in the USA. PARTICIPANTS Drexel University students, faculty, and staff (n=15 527) undergoing mandatory COVID-19 symptom tracking, testing and vaccinations. INTERVENTION Systematic symptom tracking and SARS-COV-2 testing starting in September 2020 and mandatory COVID-19 vaccination starting in September 2021. MAIN OUTCOMES AND MEASURES COVID-19 vaccine effectiveness modified by time since vaccination and symptoms. RESULTS Using fit-for-purpose digitally based symptom and vaccine tracking and mandatory comprehensive testing for SARS-CoV-2 infection, we estimate the time-dependent effects of vaccination, symptoms and covariates on the risk of infection with a Cox proportional hazards model based on calendar time scale. We found a strong protective effect of vaccination against symptomatic infection. However, there was strong evidence of a protective effect against infection only in the first 90 days after completed vaccination, and only against symptomatic versus asymptomatic infection. The overall estimated effect of vaccination within 30 days, including asymptomatic infections, was 37.3% (95% CI 26%, 47%). Vaccine effect modification by reported symptoms and time period was estimated, revealing the protective effect of vaccination within 90 days against symptomatic infection that varied from 90% (95% CI 84%, 94%) to 49%(95% CI -77%, 85%) across time periods. CONCLUSIONS This study is among the first to prospectively capture complete COVID-19 symptom, testing and vaccination data over a multiyear period. Overall effectiveness of the COVID-19 vaccine against subsequent infection, including transmissible asymptomatic infections, is modest and wanes after 90 days. Vaccination policies may need to take these issues into account.
Collapse
Affiliation(s)
- Lucy Robinson
- Department of Epidemiology and Biostatistics, Drexel University Dornsife School of Public Health, Philadelphia, Pennsylvania, USA
| | - Anna Feting
- Department of Epidemiology and Biostatistics, Drexel University Dornsife School of Public Health, Philadelphia, Pennsylvania, USA
| | - Isamu Isozaki
- Department of Computer Science, Drexel University, Philadelphia, Pennsylvania, USA
| | | | | | - Edward Kim
- Department of Computer Science, Drexel University, Philadelphia, Pennsylvania, USA
| | - Charles Cairns
- Drexel University, Philadelphia, Pennsylvania, USA
- College of Medicine, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
6
|
Sui Y, Kar S, Chawla B, Hoang T, Yu Y, Wallace SM, Andersen H, Berzofsky JA. Adjuvanted subunit intranasal vaccine reduces SARS-CoV-2 onward transmission in hamsters. Front Immunol 2025; 16:1514845. [PMID: 39981227 PMCID: PMC11841495 DOI: 10.3389/fimmu.2025.1514845] [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: 10/21/2024] [Accepted: 01/22/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Most COVID-19 vaccine trials have focused on recipient protection, not protection of their contacts, a critical need. As a subunit intranasal COVID-19 vaccine reduced nasopharyngeal virus more than did an intramuscular (IM) vaccine, we hypothesized that this vaccine might reduce onward transmission to others. Methods We vaccinated hamsters with either the IM-administrated licensed mRNA vaccine twice or one dose of mRNA IM followed by adjuvanted subunit intranasal vaccine. 24 hours after SARS-CoV-2 challenge, these animals were housed with naïve recipients in a contactless chamber that allows airborne transmission. Results Onward airborne transmission was profoundly blocked: the donor and recipients of the intranasal vaccine-boosted group had lower oral and lung viral loads (VL), which correlated with mucosal ACE2 inhibition activity. Notably, in this head-to-head comparison of COVID-19 booster vaccines on SARS-CoV-2 onward transmission, we found that statistically significant viral reduction in the lung tissues and oral swabs was observed only in the intranasal S1 nanoparticle vaccine-boosted group, but not in the systemic mRNA vaccine-boosted group, suggesting the superior protection of this intranasal vaccine, which could act as an attractive vaccine booster candidate to complement the current licensed systemic vaccines. Discussion Overall, our study strongly supports the use of the intranasal vaccine as a boost to protect not only the vaccinated person, but also people exposed to the vaccinated person, a key public health goal.
Collapse
Affiliation(s)
- Yongjun Sui
- Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | | | | | - Tanya Hoang
- Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - YuanKai Yu
- Cancer Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | | | | | - Jay A. Berzofsky
- Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
7
|
Maison DP, Tasissa H, Deitchman A, Peluso MJ, Deng Y, Miller FD, Henrich TJ, Gerschenson M. COVID-19 clinical presentation, management, and epidemiology: a concise compendium. Front Public Health 2025; 13:1498445. [PMID: 39957982 PMCID: PMC11826932 DOI: 10.3389/fpubh.2025.1498445] [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: 09/27/2024] [Accepted: 01/21/2025] [Indexed: 02/18/2025] Open
Abstract
Coronavirus Disease 2019, caused by severe acute respiratory coronavirus 2, has been an ever-evolving disease and pandemic, profoundly impacting clinical care, drug treatments, and understanding. In response to this global health crisis, there has been an unprecedented increase in research exploring new and repurposed drugs and advancing available clinical interventions and treatments. Given the widespread interest in this topic, this review aims to provide a current summary-for interested professionals not specializing in COVID-19-of the clinical characteristics, recommended treatments, vaccines, prevention strategies, and epidemiology of COVID-19. The review also offers a historical perspective on the pandemic to enhance understanding.
Collapse
Affiliation(s)
- David P. Maison
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Hawi Tasissa
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Amelia Deitchman
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Michael J. Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - F. DeWolfe Miller
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Timothy J. Henrich
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| |
Collapse
|
8
|
Manoha C, Dequiedt AL, Thery L, Marotel M, Pez F, Vouillon B, Gueneau E, de Rougemont A. Multisite community-scale monitoring of respiratory and enteric viruses in the effluent of a nursing home and in the inlet of the local wastewater treatment plant. Appl Environ Microbiol 2024; 90:e0115824. [PMID: 39387558 PMCID: PMC11577779 DOI: 10.1128/aem.01158-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Abstract
The aim of this study was to evaluate whether community-level monitoring of respiratory and enteric viruses in wastewater can provide a comprehensive picture of local virus circulation. Wastewater samples were collected weekly at the wastewater treatment plant (WWTP) inlet and at the outlet of a nearby nursing home (NH) in Burgundy, France, during the winter period of 2022/2023. We searched for the pepper mild mottle virus as an indicator of fecal content as well as for the main respiratory viruses [severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza, and respiratory syncytial virus] and enteric viruses (rotavirus, sapovirus, norovirus, astrovirus, and adenovirus). Samples were analyzed using real-time reverse transcription PCR-based methods. SARS-CoV-2 was the most frequently detected respiratory virus, with 66.7% of positive samples from the WWTP and 28.6% from the NH. Peaks of SARS-CoV-2 were consistent with the chronological incidence of infections recorded in the sentinel surveillance and the nearby hospital databases. The number of positive samples was lower in the NH than in WWTP for the three respiratory viruses. Enteric viruses were frequently detected, most often sapovirus and norovirus genogroup II, accounting both for 77.8% of positive samples in the WWTP and 57.1% and 37%, respectively, in the NH. The large circulation of sapovirus was unexpected in particular in the NH. Combined wastewater surveillance using simple optimized methods can be a valuable tool for monitoring viral circulation and may serve as a suitable early warning system for identifying both local outbreaks and the onset of epidemics. These results encourage the application of wastewater-based surveillance (WBS) to SARS-CoV2, norovirus, and sapovirus.IMPORTANCEWBS provides valuable information on the spread of epidemic viruses in the environment using appropriate and sensitive detection methods. By monitoring the circulation of viruses using reverse transcription PCR methods in wastewater from the inlet of a wastewater treatment plant and the outlet of a nearby retirement home (connected to the same collective sewer network), we aimed to demonstrate that implementing combined WBS at key community sites allows effective detection of the occurrence of respiratory (influenza, respiratory syncytial virus, and SARS-CoV-2) and enteric (norovirus, rotavirus, and sapovirus) virus infections within a given population. This analysis on a localized scale provided new information on the viral circulation in the two different sites. Implementing WBS to monitor the circulation or the emergence of infectious diseases is an important means of alerting the authorities and improving public health management. WBS could participate actively to the health of humans, animals, and the environment.
Collapse
Affiliation(s)
- Catherine Manoha
- Laboratory of Virology, University Hospital of Dijon Bourgogne, Dijon, France
| | - Anne-Laure Dequiedt
- Departmental Laboratory of Côte d’Or, Departmental Council of Côte-d'Or, Dijon, France
| | - Lucie Thery
- National Reference Centre for Gastroenteritis Viruses, University Hospital of Dijon Bourgogne, Dijon, France
| | - Marina Marotel
- Departmental Laboratory of Côte d’Or, Departmental Council of Côte-d'Or, Dijon, France
| | | | - Bruno Vouillon
- Departmental Laboratory of Côte d’Or, Departmental Council of Côte-d'Or, Dijon, France
| | - Eric Gueneau
- Departmental Laboratory of Côte d’Or, Departmental Council of Côte-d'Or, Dijon, France
| | - Alexis de Rougemont
- Laboratory of Virology, University Hospital of Dijon Bourgogne, Dijon, France
- National Reference Centre for Gastroenteritis Viruses, University Hospital of Dijon Bourgogne, Dijon, France
| |
Collapse
|
9
|
Rhodes P, Parry PI. Pharmaceutical product recall and educated hesitancy towards new drugs and novel vaccines. INTERNATIONAL JOURNAL OF RISK & SAFETY IN MEDICINE 2024; 35:317-333. [PMID: 39973420 DOI: 10.1177/09246479241292008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background: Of many pharmaceutical products launched for the benefit of humanity, a significant number have had to be recalled from the marketplace due to adverse events. A systematic review found market recalls for 462 pharmaceutical products between 1953 and 2013. In our current and remarkable period of medical history, excess mortality figures are high in many countries. Yet these statistics receive limited attention, often ignored or dismissed by mainstream news outlets. This excess mortality may include adverse effects caused by novel pharmaceutical agents that use gene-code technology.Objective: To examine key pharmaceutical product withdrawals and derive lessons that inform the current use of gene-based COVID-19 vaccines.Methods: Selective narrative review of historical pharmaceutical recalls and comparative issues with recent COVID-19 vaccines.Results: Parallels with past drug withdrawals and gene-based vaccines include distortion of clinical trial data, with critical adverse event data absent from high-impact journal publications. Delayed regulatory action on pharmacovigilance data to trigger market withdrawal occurred with Vioxx (rofecoxib) and is apparent with the gene-based COVID-19 vaccines.Conclusion: Public health requires access to raw clinical trial data, improved transparency from corporations and heightened, active pharmacovigilance worldwide.
Collapse
Affiliation(s)
- Peter Rhodes
- Gonville & Caius College, University of Cambridge, Cambridge, UK
- Anaesthesia and Intensive Care Medicine, Brisbane, QLD, Australia
| | - Peter I Parry
- Childrens Health Queensland Clinical Unit, Faculty of Medicine, University of Queensland, South Brisbane, QLD, Australia
- Department of Psychiatry, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| |
Collapse
|
10
|
Okeke KI, Ahamefule CS, Nnabuife OO, Orabueze IN, Iroegbu CU, Egbe KA, Ike AC. Antiseptics: An expeditious third force in the prevention and management of coronavirus diseases. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100293. [PMID: 39497935 PMCID: PMC11532748 DOI: 10.1016/j.crmicr.2024.100293] [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] [Indexed: 11/07/2024] Open
Abstract
Notably, severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS) and coronavirus disease 2019 (COVID-19) have all had significant negative impact on global health and economy. COVID-19 alone, has resulted to millions of deaths with new cases and mortality still being reported in its various waves. The development and use of vaccines have not stopped the transmission of SARS coronavirus 2 (SARS-CoV-2), the etiological agent of COVID-19, even among vaccinated individuals. The use of vaccines and curative drugs should be supplemented with adoption of simple hygiene preventive measures in the fight against the spread of the virus, especially for healthcare workers. Several virucidal topical antiseptics, such as povidone-iodine (PVP-I), citrox, cyclodextrins among others, have been demonstrated to be efficacious in the inactivation of SARS-CoV-2 and other coronaviruses in both in vitro and in vivo studies. The strategic application of these virucidal formulations could provide the additional impetus needed to effectively control the spread of the virus. We have here presented a simple dimension towards curtailing the dissemination of COVID-19, and other coronaviruses, through the application of effective oral, nasal and eye antiseptics among patients and medical personnel. We have further discussed the mechanism of action of some of these commonly available virucidal solutions while also highlighting some essential controversies in their use.
Collapse
Affiliation(s)
- Kizito I. Okeke
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001 Enugu State, Nigeria
| | - Chukwuemeka Samson Ahamefule
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001 Enugu State, Nigeria
| | - Obianuju O. Nnabuife
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001 Enugu State, Nigeria
| | - Ibuchukwu N. Orabueze
- Department of Medical Microbiology, University of Nigeria Teaching Hospital Enugu, Enugu State, Nigeria
| | - Christian U. Iroegbu
- Department of Microbiology, Cross River University of Technology, Calabar, Cross River State, Nigeria
| | - Kingsley A. Egbe
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001 Enugu State, Nigeria
| | - Anthony C. Ike
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001 Enugu State, Nigeria
| |
Collapse
|
11
|
Wimalawansa SJ. Unlocking insights: Navigating COVID-19 challenges and Emulating future pandemic Resilience strategies with strengthening natural immunity. Heliyon 2024; 10:e34691. [PMID: 39166024 PMCID: PMC11334859 DOI: 10.1016/j.heliyon.2024.e34691] [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: 03/06/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 08/22/2024] Open
Abstract
The original COVID-19 vaccines, developed against SARS-CoV-2, initially mitigated hospitalizations. Bivalent vaccine boosters were used widely during 2022-23, but the outbreaks persisted. Despite this, hospitalizations, mortality, and outbreaks involving dominant mutants like Alpha and Delta increased during winters when the population's vitamin D levels were at their lowest. Notably, 75 % of human immune cell/system functions, including post-vaccination adaptive immunity, rely on adequate circulatory vitamin D levels. Consequently, hypovitaminosis compromises innate and adaptive immune responses, heightening susceptibility to infections and complications. COVID-19 vaccines primarily target SARS-CoV-2 Spike proteins, thus offering only a limited protection through antibodies. mRNA vaccines, such as those for COVID-19, fail to generate secretory/mucosal immunity-like IgG responses, rendering them ineffective in halting viral spread. Additionally, mutations in the SARS-CoV-2 binding domain reduce immune recognition by vaccine-derived antibodies, leading to immune evasion by mutant viruses like Omicron variants. Meanwhile, the repeated administration of bivalent boosters intended to enhance efficacy resulted in the immunoparesis of recipients. As a result, relying solely on vaccines for outbreak prevention, it became less effective. Dominant variants exhibit increased affinity to angiotensin-converting enzyme receptor-2, enhancing infectivity but reducing virulence. Meanwhile, spike protein-related viral mutations do not impact the potency of widely available, repurposed early therapies, like vitamin D and ivermectin. With the re-emergence of COVID-19 and impending coronaviral pandemics, regulators and health organizations should proactively consider approval and strategic use of cost-effective adjunct therapies mentioned above to counter the loss of vaccine efficacy against emerging variants and novel coronaviruses and eliminate vaccine- and anti-viral agents-related serious adverse effects. Timely implementation of these strategies could reduce morbidity, mortality, and healthcare costs and provide a rational approach to address future epidemics and pandemics. This perspective critically reviews relevant literature, providing insights, justifications, and viewpoints into how the scientific community and health authorities can leverage this knowledge cost-effectively.
Collapse
Affiliation(s)
- Sunil J. Wimalawansa
- Medicine, Endocrinology, and Nutrition, B14 G2, De Soyza Flats, Moratuwa, Sri Lanka
| |
Collapse
|
12
|
Hofmeyer KA, Ventura CL, Armstrong KL, Houchens CR, Patel S, Disbrow GL, Johnson RA. Project NextGen: Developing the Next Generation of COVID-19 Vaccines and Therapeutics to Respond to the Present and Prepare for the Future. Clin Infect Dis 2024; 79:115-121. [PMID: 38356144 PMCID: PMC11259220 DOI: 10.1093/cid/ciae073] [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: 11/01/2023] [Revised: 01/03/2024] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) epidemiology and product landscapes have changed considerably since onset of the pandemic. Safe and effective vaccines and therapeutics are available, but the continual emergence of severe acute respiratory syndrome coronavirus 2 variants introduce limitations in our ability to prevent and treat disease. Project NextGen is a collaboration between the Biomedical Advanced Research and Development Authority, part of the Administration for Strategic Preparedness and Response, and the National Institute of Allergy and Infectious Diseases, part of the National Institutes of Health, that is leveraging public-private partnerships to address gaps in the nation's COVID-19 vaccine and therapeutic capabilities. Targeted investments will advance promising next-generation candidates through the most difficult phases of clinical development to encourage further private sector interest for later stage development and commercial availability. New commercial vaccines and therapeutics that are more durable and effective across variants will improve our fight against COVID-19 and transform our response to future threats.
Collapse
Affiliation(s)
- Kimberly A Hofmeyer
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Christy L Ventura
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Kimberly L Armstrong
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Christopher R Houchens
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Sandeep Patel
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Gary L Disbrow
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Robert A Johnson
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| |
Collapse
|
13
|
Junger N, Hirsch O. Ethics of Nudging in the COVID-19 Crisis and the Necessary Return to the Principles of Shared Decision Making: A Critical Review. Cureus 2024; 16:e57960. [PMID: 38601812 PMCID: PMC11005480 DOI: 10.7759/cureus.57960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 04/12/2024] Open
Abstract
Nudging, a controversial technique for modifying people's behavior in a predictable way, is claimed to preserve freedom of choice while simultaneously influencing it. Nudging had been largely confined to situations such as promoting healthy eating choices but has been employed in the coronavirus disease 2019 (COVID-19) crisis in a shift towards measures that involve significantly less choice, such as shoves and behavioral prods. Shared decision making (SDM), a method for direct involvement and autonomy, is an alternative approach to communicate risk. Predominantly peer-reviewed scientific publications from standard literature databases like PubMed, PsycInfo, and Psyndex were evaluated in a narrative review. The so-called fear nudges, as well as the dissemination of strongly emotionalizing or moralizing messages can lead to intense psycho-physical stress. The use of these nudges by specialized units during the COVID-19 pandemic generated a societal atmosphere of fear that precipitated a deterioration of the mental and physical health of the population. Major recommendations of the German COVID-19 Snapshot Monitoring (COSMO) study, which are based on elements of nudging and coercive measures, do not comply with ethical principles, basic psychological principles, or evidence-based data. SDM was misused in the COVID-19 crisis, which helped to achieve one-sided goals of governments. The emphasis on utilitarian thinking is criticized and the unethical behavior of decision makers is explained by both using the concept of moral disengagement and the maturity level of coping strategies. There should be a return to an open-ended, democratic, and pluralistic scientific debate without using nudges. It is therefore necessary to return to the origins of SDM.
Collapse
Affiliation(s)
- Nancy Junger
- Psychology, Independent Researcher, Tübingen, DEU
| | - Oliver Hirsch
- Psychology, FOM University of Applied Sciences, Siegen, DEU
| |
Collapse
|
14
|
Carducci A, Federigi I, Lauretani G, Muzio S, Pagani A, Atomsa NT, Verani M. Critical Needs for Integrated Surveillance: Wastewater-Based and Clinical Epidemiology in Evolving Scenarios with Lessons Learned from SARS-CoV-2. FOOD AND ENVIRONMENTAL VIROLOGY 2024; 16:38-49. [PMID: 38168848 DOI: 10.1007/s12560-023-09573-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024]
Abstract
During the COVID-19 pandemic, wastewater-based epidemiology (WBE) and clinical surveillance have been used as tools for analyzing the circulation of SARS-CoV-2 in the community, but both approaches can be strongly influenced by some sources of variability. From the challenging perspective of integrating environmental and clinical data, we performed a correlation analysis between SARS-CoV-2 concentrations in raw sewage and incident COVID-19 cases in areas served by medium-size wastewater treatment plants (WWTPs) from 2021 to 2023. To this aim, both datasets were adjusted for several sources of variability: WBE data were adjusted for factors including the analytical protocol, sewage flow, and population size, while clinical data adjustments considered the demographic composition of the served population. Then, we addressed the impact on the correlation of differences among sewerage networks and variations in the frequency and type of swab tests due to changes in political and regulatory scenarios. Wastewater and clinical data were significantly correlated when restrictive containment measures and limited movements were in effect (ρ = 0.50) and when COVID-19 cases were confirmed exclusively through molecular testing (ρ = 0.49). Moreover, a positive (although weak) correlation arose for WWTPs located in densely populated areas (ρ = 0.37) and with shorter sewerage lengths (ρ = 0.28). This study provides methodological approaches for interpreting WBE and clinical surveillance data, which could also be useful for other infections. Data adjustments and evaluation of possible sources of bias need to be carefully considered from the perspective of integrated environmental and clinical surveillance of infections.
Collapse
Affiliation(s)
- Annalaura Carducci
- Laboratory of Hygiene and Environmental Virology, Department of Biology, University of Pisa, Via S. Zeno 35/39, 56127, Pisa, Italy
| | - Ileana Federigi
- Laboratory of Hygiene and Environmental Virology, Department of Biology, University of Pisa, Via S. Zeno 35/39, 56127, Pisa, Italy.
| | - Giulia Lauretani
- Laboratory of Hygiene and Environmental Virology, Department of Biology, University of Pisa, Via S. Zeno 35/39, 56127, Pisa, Italy
| | - Sara Muzio
- Laboratory of Hygiene and Environmental Virology, Department of Biology, University of Pisa, Via S. Zeno 35/39, 56127, Pisa, Italy
| | - Alessandra Pagani
- Laboratory of Hygiene and Environmental Virology, Department of Biology, University of Pisa, Via S. Zeno 35/39, 56127, Pisa, Italy
| | - Nebiyu Tariku Atomsa
- Laboratory of Hygiene and Environmental Virology, Department of Biology, University of Pisa, Via S. Zeno 35/39, 56127, Pisa, Italy
| | - Marco Verani
- Laboratory of Hygiene and Environmental Virology, Department of Biology, University of Pisa, Via S. Zeno 35/39, 56127, Pisa, Italy
| |
Collapse
|
15
|
Kampf G. Does COVID-19 Vaccination Protect Contact Persons? A Systematic Review. HYGIENE 2024; 4:23-48. [DOI: 10.3390/hygiene4010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
The protective effect of COVID-19 vaccination for contact persons is controversial. Therefore, the aim of this review was to determine whether COVID-19 vaccination provides significant protection for them. A PubMed search was carried out using the terms “unvaccinated vaccinated covid” in combination with “viral load” and “transmission”. Studies were included if they reported original comparative data on the SARS-CoV-2 viral load, duration of SARS-CoV-2 detection, or SARS-CoV-2 transmission rates. A total of 332 articles were identified, of which 68 were included and analyzed. The differences in the viral load were equivocal in 57% of the 35 studies, significantly lower in the vaccinated in 11 studies and in the unvaccinated in 3 studies. The infectious virus levels were significantly lower in the vaccinated in two out of six studies. Virus clearance was significantly faster in vaccinated subjects in two of eight studies (detection of viral RNA) and two of four studies (detection of infectious virus). The secondary attack rates were significantly lower in vaccinated index cases in 6 of 15 studies. The vaccination status of contacts was described in two of the six studies and was 31.8% and 39.9% lower in households with an unvaccinated index case. The inconsistent and variable differences in the viral load, viral clearance and secondary attack rates between vaccinated and unvaccinated individuals, especially during the omicron predominance, suggests that COVID-19 vaccination is unlikely to prevent a relevant proportion of transmissions to contact persons, taking into account the relevance of the immunological status of the contact population (vaccination rates and previous infection).
Collapse
Affiliation(s)
- Günter Kampf
- University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, 17475 Greifswald, Germany
| |
Collapse
|
16
|
Wells AU. COVID-19 Vaccine Efficacy Over Time: Severe Disease in Hospitalized Patients. Radiology 2024; 310:e233340. [PMID: 38259212 DOI: 10.1148/radiol.233340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Affiliation(s)
- Athol U Wells
- From the Interstitial Lung Disease Unit, Royal Brompton and Harefield NHS Foundation Trust, London & National Lung & Lung Institute, Imperial College London, Sydney Street, London SW3 6NP, United Kingdom
| |
Collapse
|
17
|
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
|
18
|
Hijano DR, Ferrolino JA, Gu Z, Brazelton JN, Zhu H, Suganda S, Glasgow HL, Dallas RH, Allison KJ, Maron G, Darji H, Tang L, Fabrizio TP, Webby RJ, Hayden RT. Digital PCR to Measure SARS-CoV-2 RNA, Variants, and Outcomes in Youth. J Pediatric Infect Dis Soc 2023; 12:618-626. [PMID: 37956414 PMCID: PMC10725239 DOI: 10.1093/jpids/piad101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/10/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND The role of SARS-CoV-2 viral load in predicting contagiousness, disease severity, transmissibility, and clinical decision-making continues to be an area of great interest. However, most studies have been in adults and have evaluated SARS-CoV-2 loads using cycle thresholds (Ct) values, which are not standardized preventing consistent interpretation critical to understanding clinical impact and utility. Here, a quantitative SARS-CoV-2 reverse-transcription digital PCR (RT-dPCR) assay normalized to WHO International Units was applied to children at risk of severe disease diagnosed with COVID-19 at St. Jude Children's Research Hospital between March 28, 2020, and January 31, 2022. METHODS Demographic and clinical information from children, adolescents, and young adults treated at St. Jude Children's Research Hospital were abstracted from medical records. Respiratory samples underwent SARS-CoV-2 RNA quantitation by RT-dPCR targeting N1 and N2 genes, with sequencing to determine the genetic lineage of infecting virus. RESULTS Four hundred and sixty-two patients aged 0-24 years (median 11 years old) were included during the study period. Most patients were infected by the omicron variant (43.72%), followed by ancestral strain (22.29%), delta (13.20%), and alpha (2.16%). Viral load at presentation ranged from 2.49 to 9.14 log10 IU/mL, and higher viral RNA loads were associated with symptoms (OR 1.32; CI 95% 1.16-1.49) and respiratory disease (OR 1.23; CI 95% 1.07-1.41). Viral load did not differ by SARS-CoV-2 variant, vaccination status, age, or baseline diagnosis. CONCLUSIONS SARS-CoV-2 RNA loads predict the presence of symptomatic and respiratory diseases. The use of standardized, quantitative methods is feasible, allows for replication, and comparisons across institutions, and has the potential to facilitate consensus quantitative thresholds for risk stratification and treatment.
Collapse
Affiliation(s)
- Diego R Hijano
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jose A Ferrolino
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Zhengming Gu
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jessica N Brazelton
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Haiqing Zhu
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Sri Suganda
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Heather L Glasgow
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Ronald H Dallas
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Kim J Allison
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Gabriela Maron
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Himani Darji
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Li Tang
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Thomas P Fabrizio
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Richard J Webby
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Randall T Hayden
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
19
|
Fundora MP, Kamidani S, Oster ME. COVID Vaccination as a Strategy for Cardiovascular Disease Prevention. Curr Cardiol Rep 2023; 25:1327-1335. [PMID: 37688764 DOI: 10.1007/s11886-023-01950-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/21/2023] [Indexed: 09/11/2023]
Abstract
PURPOSE OF REVIEW Cardiovascular (CV) disease is a known complication of SARS-CoV-2 infection. A clear benefit of COVID-19 vaccination is a reduction mortality; however, COVID-19 vaccination may also prevent cardiovascular disease (CVD). We aim to describe CV pathology associated with SARS-CoV-2 infection and describe how COVID-19 vaccination is a strategy for CVD prevention. RECENT FINDINGS The risks and benefits of COVID-19 vaccination have been widely studied. Analysis of individuals with and without pre-existing CVD has shown that COVID-19 vaccination can prevent morbidity associated with SARS-CoV-2 infection and reduce mortality. COVID-19 vaccination is effective in preventing myocardial infarction, cerebrovascular events, myopericarditis, and long COVID, all associated with CVD risk factors. Vaccination reduces mortality in patients with pre-existing CVD. Further study investigating ideal vaccination schedules for individuals with CVD should be undertaken to protect this vulnerable group and address new risks from variants of concern.
Collapse
Affiliation(s)
- Michael P Fundora
- Children's Healthcare of Atlanta Cardiology, Department of Pediatrics, Emory University, 1405 Clifton Rd NE, Atlanta, GA, 30322, USA
| | - Satoshi Kamidani
- The Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and the Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Matthew E Oster
- Children's Healthcare of Atlanta Cardiology, Department of Pediatrics, Emory University, 1405 Clifton Rd NE, Atlanta, GA, 30322, USA.
| |
Collapse
|
20
|
Longo BM, Venuti F, Gaviraghi A, Lupia T, Ranzani FA, Pepe A, Ponzetta L, Vita D, Allice T, Gregorc V, Frascione PMM, De Rosa FG, Calcagno A, Bonora S. Sequential or Combination Treatments as Rescue Therapies in Immunocompromised Patients with Persistent SARS-CoV-2 Infection in the Omicron Era: A Case Series. Antibiotics (Basel) 2023; 12:1460. [PMID: 37760757 PMCID: PMC10525462 DOI: 10.3390/antibiotics12091460] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Prolonged SARS-CoV-2 infections are widely described in immunosuppressed patients, but safe and effective treatment strategies are lacking. We aimed to outline our approach to treating persistent COVID-19 in patients with immunosuppression from different causes. In this case series, we retrospectively enrolled all immunosuppressed patients with persistent SARS-CoV-2 infections treated at our centers between March 2022 and February 2023. Patients received different sequential or combination regimens, including antivirals (remdesivir, nirmatrelvir/ritonavir, or molnupiravir) and/or monoclonal antibodies (mAbs) (tixagevimab/cilgavimab or sotrovimab). The main outcome was a complete virological response (negative SARS-CoV-2 RT-PCR on nasopharyngeal swabs) at the end of treatment. Fifteen patients were included as follows: eleven (11/15; 73%) with hematological disease and four (4/15; 27%) with recently diagnosed HIV/AIDS infection. Six patients (6/15; 40%) received a single antiviral course, four patients (4/15; 27%) received an antiviral and mAbs sequentially, and two patients (13%) received three lines of treatment (a sequence of three antivirals or two antivirals and mAbs). A combination of two antivirals or one antiviral plus mAbs was administered in three cases (3/15, 20%). One patient died while still positive for SARS-CoV-2, while fourteen (14/15; 93%) tested negative within 16 days after the end of treatment. The median time to negativization since the last treatment was 2.5 days. Both sequential and combination regimens used in this study demonstrated high efficacy and safety in the high-risk group of immunosuppressed patients.
Collapse
Affiliation(s)
- Bianca Maria Longo
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, “Amedeo di Savoia” Hospital, ASL “Città di Torino”, 10060 Turin, Italy (F.V.); (D.V.); (F.G.D.R.)
| | - Francesco Venuti
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, “Amedeo di Savoia” Hospital, ASL “Città di Torino”, 10060 Turin, Italy (F.V.); (D.V.); (F.G.D.R.)
| | - Alberto Gaviraghi
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, “Amedeo di Savoia” Hospital, ASL “Città di Torino”, 10060 Turin, Italy (F.V.); (D.V.); (F.G.D.R.)
| | - Tommaso Lupia
- Unit of Infectious Diseases, Cardinal Massaia Hospital, 14100 Asti, Italy
| | - Fabio Antonino Ranzani
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, “Amedeo di Savoia” Hospital, ASL “Città di Torino”, 10060 Turin, Italy (F.V.); (D.V.); (F.G.D.R.)
| | - Andrea Pepe
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, “Amedeo di Savoia” Hospital, ASL “Città di Torino”, 10060 Turin, Italy (F.V.); (D.V.); (F.G.D.R.)
| | - Laura Ponzetta
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, “Amedeo di Savoia” Hospital, ASL “Città di Torino”, 10060 Turin, Italy (F.V.); (D.V.); (F.G.D.R.)
| | - Davide Vita
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, “Amedeo di Savoia” Hospital, ASL “Città di Torino”, 10060 Turin, Italy (F.V.); (D.V.); (F.G.D.R.)
| | - Tiziano Allice
- Microbiology and Molecular Biology Laboratory, “Amedeo di Savoia” Hospital, ASL “Città di Torino”, 10060 Turin, Italy;
| | - Vanesa Gregorc
- Unit of Oncology and Haematology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
| | | | - Francesco Giuseppe De Rosa
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, “Amedeo di Savoia” Hospital, ASL “Città di Torino”, 10060 Turin, Italy (F.V.); (D.V.); (F.G.D.R.)
- Department of Medical Sciences, Infectious Diseases, University of Turin, 10126 Turin, Italy
| | - Andrea Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, “Amedeo di Savoia” Hospital, ASL “Città di Torino”, 10060 Turin, Italy (F.V.); (D.V.); (F.G.D.R.)
| | - Stefano Bonora
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, “Amedeo di Savoia” Hospital, ASL “Città di Torino”, 10060 Turin, Italy (F.V.); (D.V.); (F.G.D.R.)
| |
Collapse
|
21
|
Chen HF, Wang WJ, Chen CY, Chang WC, Hsueh PR, Peng SL, Wu CS, Chen Y, Huang HY, Shen WJ, Wang SC, Hung MC. The natural tannins oligomeric proanthocyanidins and punicalagin are potent inhibitors of infection by SARS-CoV-2. eLife 2023; 12:e84899. [PMID: 37642993 PMCID: PMC10465125 DOI: 10.7554/elife.84899] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 07/17/2023] [Indexed: 08/31/2023] Open
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic continues to infect people worldwide. While the vaccinated population has been increasing, the rising breakthrough infection persists in the vaccinated population. For living with the virus, the dietary guidelines to prevent virus infection are worthy of and timely to develop further. Tannic acid has been demonstrated to be an effective inhibitor of coronavirus and is under clinical trial. Here we found that two other members of the tannins family, oligomeric proanthocyanidins (OPCs) and punicalagin, are also potent inhibitors against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection with different mechanisms. OPCs and punicalagin showed inhibitory activity against omicron variants of SARS-CoV-2 infection. The water extractant of the grape seed was rich in OPCs and also exhibited the strongest inhibitory activities for viral entry of wild-type and other variants in vitro. Moreover, we evaluated the inhibitory activity of grape seed extractants (GSE) supplementation against SARS-CoV-2 viral entry in vivo and observed that serum samples from the healthy human subjects had suppressive activity against different variants of SARS-CoV-2 Vpp infection after taking GSE capsules. Our results suggest that natural tannins acted as potent inhibitors against SARS-CoV-2 infection, and GSE supplementation could serve as healthy food for infection prevention.
Collapse
Affiliation(s)
- Hsiao-Fan Chen
- Graduate Institute of Biomedical Sciences, China Medical UniversityTaichungTaiwan
- Research Center for Cancer Biology, China Medical UniversityTaichungTaiwan
| | - Wei-Jan Wang
- Research Center for Cancer Biology, China Medical UniversityTaichungTaiwan
- Department of Biological Science and Technology, College of Life Sciences, China Medical UniversityTaichungTaiwan
| | - Chung-Yu Chen
- Research Center for Cancer Biology, China Medical UniversityTaichungTaiwan
| | - Wei-Chao Chang
- Center for Molecular Medicine, China Medical University Hospital, China Medical UniversityTaichungTaiwan
| | - Po-Ren Hsueh
- Departments of Laboratory Medicine and Internal Medicine, China Medical University Hospital, School of Medicine, China Medical UniversityTaichungTaiwan
| | - Shin-Lei Peng
- Department of Biomedical Imaging and Radiological Science, China Medical UniversityTaichungTaiwan
- Neuroscience and Brain Disease Center, China Medical UniversityTaichungTaiwan
| | - Chen-Shiou Wu
- Graduate Institute of Biomedical Sciences, China Medical UniversityTaichungTaiwan
- Research Center for Cancer Biology, China Medical UniversityTaichungTaiwan
| | - Yeh Chen
- Department of Biological Science and Technology, College of Life Sciences, China Medical UniversityTaichungTaiwan
- Institute of New Drug Development, China Medical UniversityTaichungTaiwan
| | - Hsin-Yu Huang
- Graduate Institute of Biomedical Sciences, China Medical UniversityTaichungTaiwan
| | - Wan-Jou Shen
- Graduate Institute of Biomedical Sciences, China Medical UniversityTaichungTaiwan
| | - Shao-Chun Wang
- Graduate Institute of Biomedical Sciences, China Medical UniversityTaichungTaiwan
- Research Center for Cancer Biology, China Medical UniversityTaichungTaiwan
- Center for Molecular Medicine, China Medical University Hospital, China Medical UniversityTaichungTaiwan
- Cancer Biology and Precision Therapeutics Center, China Medical UniversityTaichungTaiwan
- Department of Biotechnology, Asia UniversityTaichungTaiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, China Medical UniversityTaichungTaiwan
- Research Center for Cancer Biology, China Medical UniversityTaichungTaiwan
- Center for Molecular Medicine, China Medical University Hospital, China Medical UniversityTaichungTaiwan
- Cancer Biology and Precision Therapeutics Center, China Medical UniversityTaichungTaiwan
- Department of Biotechnology, Asia UniversityTaichungTaiwan
- Institute of Biochemistry and Molecular Biology, China Medical UniversityTaichungTaiwan
| |
Collapse
|
22
|
Tukhvatulin AI, Dolzhikova IV, Dzharullaeva AS, Grousova DM, Kovyrshina AV, Zubkova OV, Zorkov ID, Iliukhina AA, Shelkov AY, Erokhova AS, Popova O, Ozharovskaia TA, Zrelkin DI, Izhaeva FM, Shcheblyakov DV, Esmagambetov IB, Tokarskaya EA, Nikitenko NA, Lubenets NL, Khadorich EA, Gushchin VA, Borzakova SN, Vlasova AV, Osmanov IM, Gorev VV, Naroditsky BS, Logunov DY, Gintsburg AL. Safety and immunogenicity of rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine against SARS-CoV-2 in healthy adolescents: an open-label, non-randomized, multicenter, phase 1/2, dose-escalation study. Front Immunol 2023; 14:1228461. [PMID: 37600800 PMCID: PMC10432829 DOI: 10.3389/fimmu.2023.1228461] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/11/2023] [Indexed: 08/22/2023] Open
Abstract
To protect young individuals against SARS-CoV-2 infection, we conducted an open-label, prospective, non-randomised dose-escalation Phase 1/2 clinical trial to evaluate the immunogenicity and safety of the prime-boost "Sputnik V" vaccine administered at 1/10 and 1/5 doses to adolescents aged 12-17 years. The study began with the vaccination of the older cohort (15-to-17-year-old participants) with the lower (1/10) dose of vaccine and then expanded to the whole group (12-to-17-year-old participants). Next, 1/5 dose was used according to the same scheme. Both doses were well tolerated by all age groups. No serious or severe adverse events were detected. Most of the solicited adverse reactions were mild. No significant differences in total frequencies of adverse events were registered between low and high doses in age-pooled groups (69.6% versus 66.7%). In contrast, the 1/5 dose induced significantly higher humoral and T cell-mediated immune responses than the 1/10 dose. The 1/5 vaccine dose elicited higher antigen-binding (both S and RBD-specific) as well as virus-neutralising antibody titres at the maximum of response (day 42), also resulting in a statistically significant difference at a distanced timepoint (day 180) compared to the 1/10 vaccine dose. Higher dose resulted in increased cross-neutralization of Delta and Omicron variants. Clinical Trial Registration ClinicalTrials.gov, NCT04954092, LP-007632.
Collapse
Affiliation(s)
- Amir I. Tukhvatulin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Inna V. Dolzhikova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alina S. Dzharullaeva
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Daria M. Grousova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anna V. Kovyrshina
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Olga V. Zubkova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Ilya D. Zorkov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anna A. Iliukhina
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Artem Y. Shelkov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alina S. Erokhova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Olga Popova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Tatiana A. Ozharovskaia
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Denis I. Zrelkin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Fatima M. Izhaeva
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Dmitry V. Shcheblyakov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Ilias B. Esmagambetov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Elisaveta A. Tokarskaya
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Natalia A. Nikitenko
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Nadezhda L. Lubenets
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Elizaveta A. Khadorich
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladimir A. Gushchin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Svetlana N. Borzakova
- Children’s City Clinical Hospital named after Z. A. Bashlyaeva, Moscow City Health Department, Moscow, Russia
| | - Anna V. Vlasova
- Morozov Children’s City Clinical Hospital, Moscow Health Department, Moscow, Russia
| | - Ismail M. Osmanov
- Children’s City Clinical Hospital named after Z. A. Bashlyaeva, Moscow City Health Department, Moscow, Russia
| | - Valerii V. Gorev
- Morozov Children’s City Clinical Hospital, Moscow Health Department, Moscow, Russia
| | - Boris S. Naroditsky
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Denis Y. Logunov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
- Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexander L. Gintsburg
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya”, Ministry of Health of the Russian Federation, Moscow, Russia
- Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
23
|
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: 0.5] [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
|
24
|
Ravindran S, Gubbay JB, Cronin K, Sullivan A, Zygmunt A, Johnson K, Buchan SA, Parpia AS. Association Between Cycle Threshold Value and Vaccination Status Among Severe Acute Respiratory Syndrome Coronavirus 2 Omicron Variant Cases in Ontario, Canada, in December 2021. Open Forum Infect Dis 2023; 10:ofad282. [PMID: 37274182 PMCID: PMC10234392 DOI: 10.1093/ofid/ofad282] [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: 03/28/2023] [Accepted: 05/18/2023] [Indexed: 06/06/2023] Open
Abstract
Background Increased immune evasion by emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants and occurrence of breakthrough infections raise questions about whether coronavirus disease 2019 vaccination status affects SARS-CoV-2 viral load among those infected. This study examined the relationship between cycle threshold (Ct) value, which is inversely associated with viral load, and vaccination status at the onset of the Omicron wave onset in Ontario, Canada. Methods Using linked provincial databases, we compared median Ct values across vaccination status among polymerase chain reaction-confirmed Omicron variant SARS-CoV-2 cases (sublineages B.1.1.529, BA.1, and BA.1.1) between 6 and 30 December 2021. Cases were presumed to be Omicron based on S-gene target failure. We estimated the relationship between vaccination status and Ct values using multiple linear regression, adjusting for age group, sex, and symptom status. Results Of the 27 029 presumed Omicron cases in Ontario, the majority were in individuals who had received a complete vaccine series (87.7%), followed by unvaccinated individuals (8.1%), and those who had received a booster dose (4.2%). The median Ct value for post-booster dose individuals (18.3 [interquartile range, 15.4-22.3]) was significantly higher than that for unvaccinated (17.9 [15.2-21.6]; P = .02) and post-vaccine series individuals (17.8 [15.3-21.5]; P = .005). Post-booster dose cases remained associated with a significantly higher median Ct value than cases in unvaccinated individuals (P ≤ .001), after adjustment for covariates. Compared with values in persons aged 18-29 years, Ct values were significantly lower among most age groups >50 years. Conclusions While slightly lower Ct values were observed among unvaccinated individuals infected with Omicron compared with post-booster dose cases, further research is required to determine whether a significant difference in secondary transmission exists between these groups.
Collapse
Affiliation(s)
| | - Jonathan B Gubbay
- Public Health Ontario Laboratory, Public Health Ontario, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology and Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Kirby Cronin
- Public Health Ontario Laboratory, Public Health Ontario, Toronto, Ontario, Canada
| | - Ashleigh Sullivan
- Public Health Ontario Laboratory, Public Health Ontario, Toronto, Ontario, Canada
| | - Austin Zygmunt
- Health Protection, Public Health Ontario, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology and Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | - Karen Johnson
- Health Protection, Public Health Ontario, Toronto, Ontario, Canada
| | - Sarah A Buchan
- Correspondence: Sarah A. Buchan, Public Health Ontario, 661 University Ave, Floor 17, Toronto, ON M5G 1M1, Canada (); Alyssa S. Parpia, Public Health Ontario, 480 University Ave, Toronto, ON M5G 1V2, Canada ()
| | - Alyssa S Parpia
- Correspondence: Sarah A. Buchan, Public Health Ontario, 661 University Ave, Floor 17, Toronto, ON M5G 1M1, Canada (); Alyssa S. Parpia, Public Health Ontario, 480 University Ave, Toronto, ON M5G 1V2, Canada ()
| |
Collapse
|
25
|
Fraser R, Orta-Resendiz A, Mazein A, Dockrell DH. Upper respiratory tract mucosal immunity for SARS-CoV-2 vaccines. Trends Mol Med 2023; 29:255-267. [PMID: 36764906 PMCID: PMC9868365 DOI: 10.1016/j.molmed.2023.01.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023]
Abstract
SARS-CoV-2 vaccination significantly reduces morbidity and mortality, but has less impact on viral transmission rates, thus aiding viral evolution, and the longevity of vaccine-induced immunity rapidly declines. Immune responses in respiratory tract mucosal tissues are crucial for early control of infection, and can generate long-term antigen-specific protection with prompt recall responses. However, currently approved SARS-CoV-2 vaccines are not amenable to adequate respiratory mucosal delivery, particularly in the upper airways, which could account for the high vaccine breakthrough infection rates and limited duration of vaccine-mediated protection. In view of these drawbacks, we outline a strategy that has the potential to enhance both the efficacy and durability of existing SARS-CoV-2 vaccines, by inducing robust memory responses in the upper respiratory tract (URT) mucosa.
Collapse
Affiliation(s)
- Rupsha Fraser
- The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| | - Aurelio Orta-Resendiz
- Institut Pasteur, Université Paris Cité, HIV, Inflammation and Persistence Unit, F-75015 Paris, France
| | - Alexander Mazein
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - David H Dockrell
- The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| |
Collapse
|
26
|
Cotter CA, Americo JL, Earl PL, Moss B. Protection from SARS-CoV-2 Variants by MVAs expressing matched or mismatched S administered intranasally to mice. NPJ Vaccines 2023; 8:47. [PMID: 36973267 PMCID: PMC10040904 DOI: 10.1038/s41541-023-00645-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
SARS-CoV-2 vaccines prevent severe disease but are less efficient in averting infection and transmission of variant strains, making it imperative to explore ways of enhancing protection. Use of inbred mice expressing the human SARS-CoV-2 receptor facilitates such investigations. We employed recombinant MVAs (rMVAs) expressing modified S of several SARS-CoV-2 strains and compared their ability to neutralize variants, bind S proteins and protect K18-hACE2 mice against SARS-CoV-2 challenge when administered intramuscularly or intranasally. The rMVAs expressing Wuhan, Beta and Delta S induced substantial cross neutralizing activities to each other but very low neutralization of Omicron; while rMVA expressing Omicon S induced neutralizing antibody predominanly to Omicron. In mice primed and boosted with rMVA expressing the Wuhan S, neutralizing antibodies to Wuhan increased after one immunization with rMVA expressing Omicron S due to original antigenic sin, but substantial neutralizing antibody to Omicron required a second immunization. Nevertheless, monovalent vaccines with S mismatched to the challenge virus still protected against severe disease and reduced the amounts of virus and subgenomic RNAs in the lungs and nasal turbinates, though not as well as vaccines with matched S. Passive transfer of Wuhan immune serum with Omicron S binding but undetectable neutralizing activity reduced infection of the l-ungs by Omicron suggesting additional effector functions. Notably, there was less infectious virus and viral subgenomic RNAs in the nasal turbinates and lungs when the rMVAs were administered intranasally rather than intramuscularly and this held true for vaccines that were matched or mismatched to the challenge strain of SARS-CoV-2.
Collapse
Affiliation(s)
- Catherine A Cotter
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey L Americo
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patricia L Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
27
|
Tian X, Zhang Y, Wang W, Fang F, Zhang W, Zhu Z, Wan Y. The impacts of vaccination status and host factors during early infection on SARS-CoV-2 persistence:a retrospective single-center cohort study. Int Immunopharmacol 2023; 114:109534. [PMID: 36476489 PMCID: PMC9708622 DOI: 10.1016/j.intimp.2022.109534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/13/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND Viral persistence is a crucial factor that influences the transmissibility of SARS-CoV-2. However, the impacts of vaccination and physiological variables on viral persistence have not been adequately clarified. METHODS We collected the clinical records of 377 COVID-19 patients, which contained unvaccinated patients and patients received two doses of an inactivated vaccine or an mRNA vaccine. The impacts of vaccination on disease severity and viral persistence and the correlations between 49 laboratory variables and viral persistence were analyzed separately. Finally, we established a multivariate regression model to predict the persistence of viral RNA. RESULTS Both inactivated and mRNA vaccines significantly reduced the rate of moderate cases, while the vaccine related shortening of viral RNA persistence was only observed in moderate patients. Correlation analysis showed that 10 significant laboratory variables were shared by the unvaccinated mild patients and mild patients inoculated with an inactivated vaccine, but not by the mild patients inoculated with an mRNA vaccine. A multivariate regression model established based on the variables correlating with viral persistence in unvaccinated mild patients could predict the persistence of viral RNA for all patients except three moderate patients inoculated with an mRNA vaccine. CONCLUSION Vaccination contributed limitedly to the clearance of viral RNA in COVID-19 patients. While, laboratory variables in early infection could predict the persistence of viral RNA.
Collapse
Affiliation(s)
- Xiangxiang Tian
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan Province, Zhengzhou 450052, China; Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Clinical Laboratory, The First People's Hospital of Shangqiu, Shangqiu 476000, China
| | - Yifan Zhang
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan Province, Zhengzhou 450052, China; Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wanhai Wang
- Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan Province, Zhengzhou 450052, China
| | - Fang Fang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China; State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai 200000, China
| | - Zhaoqin Zhu
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China.
| | - Yanmin Wan
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China; State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai 200000, China.
| |
Collapse
|
28
|
Asymptomatic screening for severe acute respiratory coronavirus virus 2 (SARS-CoV-2) as an infection prevention measure in healthcare facilities: Challenges and considerations. Infect Control Hosp Epidemiol 2023; 44:2-7. [PMID: 36539917 DOI: 10.1017/ice.2022.295] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Testing of asymptomatic patients for severe acute respiratory coronavirus virus 2 (SARS-CoV-2) (ie, "asymptomatic screening) to attempt to reduce the risk of nosocomial transmission has been extensive and resource intensive, and such testing is of unclear benefit when added to other layers of infection prevention mitigation controls. In addition, the logistic challenges and costs related to screening program implementation, data noting the lack of substantial aerosol generation with elective controlled intubation, extubation, and other procedures, and the adverse patient and facility consequences of asymptomatic screening call into question the utility of this infection prevention intervention. Consequently, the Society for Healthcare Epidemiology of America (SHEA) recommends against routine universal use of asymptomatic screening for SARS-CoV-2 in healthcare facilities. Specifically, preprocedure asymptomatic screening is unlikely to provide incremental benefit in preventing SARS-CoV-2 transmission in the procedural and perioperative environment when other infection prevention strategies are in place, and it should not be considered a requirement for all patients. Admission screening may be beneficial during times of increased virus transmission in some settings where other layers of controls are limited (eg, behavioral health, congregate care, or shared patient rooms), but widespread routine use of admission asymptomatic screening is not recommended over strengthening other infection prevention controls. In this commentary, we outline the challenges surrounding the use of asymptomatic screening, including logistics and costs of implementing a screening program, and adverse patient and facility consequences. We review data pertaining to the lack of substantial aerosol generation during elective controlled intubation, extubation, and other procedures, and we provide guidance for when asymptomatic screening for SARS-CoV-2 may be considered in a limited scope.
Collapse
|
29
|
Universal admission laboratory screening for severe acute respiratory coronavirus virus 2 (SARS-CoV-2) asymptomatic infection across a large health system. Infect Control Hosp Epidemiol 2023; 44:68-74. [PMID: 36533305 DOI: 10.1017/ice.2022.301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Admission laboratory screening for asymptomatic coronavirus disease 2019 (COVID-19) has been utilized to mitigate healthcare-associated severe acute respiratory coronavirus virus 2 (SARS-CoV-2) transmission. An understanding of the impact of such testing across a variety of patient populations is needed. METHODS SARS-CoV-2 nucleic acid amplification admission testing results for all asymptomatic patients across 4 distinct inpatient facilities between April 20, 2020, and June 14, 2021, were analyzed. Positivity rates and the number needed to test (NNT) to identify 1 asymptomatic infected patient were calculated. Admission results were compared to COVID-19 community incidence rates for the system's surrounding metropolitan service area. Using a national survey of hospital epidemiologists, a clinically meaningful NNT of 1:100 was identified. RESULTS In total, 51,187 tests were collected (positivity rate, 1.8%). During periods of high transmission, the NNT met the clinically relevant threshold in all populations. The NNT approached or met the threshold for most locations during periods of lower transmission. For all transmission levels, the NNT for fully vaccinated patients did not meet the threshold. CONCLUSIONS Implementing an asymptomatic patient admission testing program can provide clinically relevant data based on the NNT, even during periods of lower transmission and among different patient populations. Limiting admission testing to non-fully vaccinated patients during periods of lower transmission may be a strategy to address resource concerns around this practice. Although the impact of such testing on healthcare-associated COVID-19 among patients and healthcare workers could not be clearly determined, these data provide important information as facilities weigh the costs and benefits of such testing.
Collapse
|
30
|
Diani S, Leonardi E, Cavezzi A, Ferrari S, Iacono O, Limoli A, Bouslenko Z, Natalini D, Conti S, Mantovani M, Tramonte S, Donzelli A, Serravalle E. SARS-CoV-2-The Role of Natural Immunity: A Narrative Review. J Clin Med 2022; 11:6272. [PMID: 36362500 PMCID: PMC9655392 DOI: 10.3390/jcm11216272] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Both natural immunity and vaccine-induced immunity to COVID-19 may be useful to reduce the mortality/morbidity of this disease, but still a lot of controversy exists. AIMS This narrative review analyzes the literature regarding these two immunitary processes and more specifically: (a) the duration of natural immunity; (b) cellular immunity; (c) cross-reactivity; (d) the duration of post-vaccination immune protection; (e) the probability of reinfection and its clinical manifestations in the recovered patients; (f) the comparisons between vaccinated and unvaccinated as to the possible reinfections; (g) the role of hybrid immunity; (h) the effectiveness of natural and vaccine-induced immunity against Omicron variant; (i) the comparative incidence of adverse effects after vaccination in recovered individuals vs. COVID-19-naïve subjects. MATERIAL AND METHODS through multiple search engines we investigated COVID-19 literature related to the aims of the review, published since April 2020 through July 2022, including also the previous articles pertinent to the investigated topics. RESULTS nearly 900 studies were collected, and 246 pertinent articles were included. It was highlighted that the vast majority of the individuals after suffering from COVID-19 develop a natural immunity both of cell-mediated and humoral type, which is effective over time and provides protection against both reinfection and serious illness. Vaccine-induced immunity was shown to decay faster than natural immunity. In general, the severity of the symptoms of reinfection is significantly lower than in the primary infection, with a lower degree of hospitalizations (0.06%) and an extremely low mortality. CONCLUSIONS this extensive narrative review regarding a vast number of articles highlighted the valuable protection induced by the natural immunity after COVID-19, which seems comparable or superior to the one induced by anti-SARS-CoV-2 vaccination. Consequently, vaccination of the unvaccinated COVID-19-recovered subjects may not be indicated. Further research is needed in order to: (a) measure the durability of immunity over time; (b) evaluate both the impacts of Omicron BA.5 on vaccinated and healed subjects and the role of hybrid immunity.
Collapse
Affiliation(s)
- Sara Diani
- School of Musictherapy, Université Européenne Jean Monnet, 35129 Padova, Italy
| | | | | | | | - Oriana Iacono
- Physical Medicine and Rehabilitation Department, Mirandola Hospital, 41037 Mirandola, Italy
| | - Alice Limoli
- ARPAV (Regional Agency for the Environment Protection), 31100 Treviso, Italy
| | - Zoe Bouslenko
- Cardiology Department, Valdese Hospital, 10100 Torino, Italy
| | | | | | | | - Silvano Tramonte
- Environment and Health Commission, National Bioarchitecture Institute, 20121 Milano, Italy
| | | | | |
Collapse
|
31
|
Márquez-Alvarez V, Amigó-Vega J, Rivera A, Batista-Leyva AJ, Altshuler E. Relative assessment of cloth mask protection against ballistic droplets: A frugal approach. PLoS One 2022; 17:e0275376. [PMID: 36194594 PMCID: PMC9531801 DOI: 10.1371/journal.pone.0275376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/15/2022] [Indexed: 11/07/2022] Open
Abstract
During the COVID-19 pandemic, the relevance of evaluating the effectiveness of face masks-especially those made at home using a variety of materials-has become obvious. However, quantifying mask protection often requires sophisticated equipment. Using a frugal stain technique, here we quantify the "ballistic" droplets reaching a receptor from a jet-emitting source which mimics a coughing, sneezing or talking human-in real life, such droplets may host active SARS-CoV-2 virus able to replicate in the nasopharynx. We demonstrate that materials often used in home-made face masks block most of the droplets. Mimicking situations eventually found in daily life, we also show quantitatively that less liquid carried by ballistic droplets reaches a receptor when a blocking material is deployed near the source than when located near the receptor, which supports the paradigm that your face mask does protect you, but protects others even better than you. Finally, the blocking behavior can be quantitatively explained by a simple mechanical model.
Collapse
Affiliation(s)
| | - Joaquín Amigó-Vega
- Center for Complex Systems, Physics Faculty, University of Havana, Havana, Cuba
| | - Aramis Rivera
- Zeolite Engineering Laboratory, Institute of Materials and Reagents (IMRE), University of Havana, Havana, Cuba
| | - Alfo José Batista-Leyva
- Center for Complex Systems, Physics Faculty, University of Havana, Havana, Cuba
- Instituto Superior de Ciencia y Tecnología Aplicadas (InSTEC), University of Havana, Havana, Cuba
| | - Ernesto Altshuler
- Center for Complex Systems, Physics Faculty, University of Havana, Havana, Cuba
| |
Collapse
|
32
|
Wu F, Lee WL, Chen H, Gu X, Chandra F, Armas F, Xiao A, Leifels M, Rhode SF, Wuertz S, Thompson J, Alm EJ. Making waves: Wastewater surveillance of SARS-CoV-2 in an endemic future. WATER RESEARCH 2022; 219:118535. [PMID: 35605390 PMCID: PMC9062764 DOI: 10.1016/j.watres.2022.118535] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 05/28/2023]
Abstract
Wastewater-based surveillance (WBS) has been widely used as a public health tool to monitor the emergence and spread of SARS-CoV-2 infections in populations during the COVID-19 pandemic. Coincident with the global vaccination efforts, the world is also enduring new waves of SARS-CoV-2 variants. Reinfections and vaccine breakthroughs suggest an endemic future where SARS-CoV-2 continues to persist in the general population. In this treatise, we aim to explore the future roles of wastewater surveillance. Practically, WBS serves as a relatively affordable and non-invasive tool for mass surveillance of SARS-CoV-2 infection while minimizing privacy concerns, attributes that make it extremely suited for its long-term usage. In an endemic future, the utility of WBS will include 1) monitoring the trend of viral loads of targets in wastewater for quantitative estimate of changes in disease incidence; 2) sampling upstream for pinpointing infections in neighborhoods and at the building level; 3) integrating wastewater and clinical surveillance for cost-efficient population surveillance; and 4) genome sequencing wastewater samples to track circulating and emerging variants in the population. We further discuss the challenges and future developments of WBS to reduce inconsistencies in wastewater data worldwide, improve its epidemiological inference, and advance viral tracking and discovery as a preparation for the next viral pandemic.
Collapse
Affiliation(s)
- Fuqing Wu
- Center for Infectious Disease, Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas School of Public Health, Houston, TX, USA.
| | - Wei Lin Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | - Hongjie Chen
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | - Xiaoqiong Gu
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | - Franciscus Chandra
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | - Federica Armas
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | - Amy Xiao
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mats Leifels
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore
| | | | - Stefan Wuertz
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore; School of Civil and Environmental Engineering, Nanyang Technological University, Singapore
| | - Janelle Thompson
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore; Asian School of the Environment, Nanyang Technological University, Singapore
| | - Eric J Alm
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|