1
|
Stone EC, Okasako-Schmucker DL, Taliano J, Schaefer M, Kuhar DT. Risk period for transmission of SARS-CoV-2 and seasonal influenza: a rapid review. Infect Control Hosp Epidemiol 2025; 46:1-9. [PMID: 39989317 PMCID: PMC11883656 DOI: 10.1017/ice.2025.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 02/25/2025]
Abstract
BACKGROUND Restricting infectious healthcare workers (HCWs) from the workplace is an important infection prevention strategy. The duration of viral shedding or symptoms are often used as proxies for the infectious period in adults but may not accurately estimate it. OBJECTIVE To determine the risk period for transmission among previously healthy adults infected with SARS-CoV-2 omicron variant (omicron) or influenza A (influenza) by examining the duration of shedding and symptoms, and day of symptom onset in secondary cases of transmission pairs. DESIGN Rapid review. METHODS This rapid review adhered to PRISMA-ScR; five databases were searched. The cumulative daily proportion of participants with an outcome of interest was calculated for each study and summarized. RESULTS Forty-three studies were included. Shedding resolved among ≥ 70% of participants by the end of day nine post symptom onset for omicron, and day seven for influenza; and for ≥ 90% of participants, by the end of day 10 for omicron and day nine for influenza. Two studies suggested shedding continues > 24 hours post-fever resolution for both viruses. Symptom onset occurred in ≥ 80% of secondary cases by the end of day seven post-primary case symptom onset for omicron and day six for influenza. CONCLUSIONS Omicron shedding is consistent with previous recommendations to exclude infected HCWs from work for 10 days; and influenza follows a similar trend. Earlier symptom onset in most secondary cases for both pathogens indicates that, despite persistent viral shedding, most transmission occurs earlier; and the cumulative serial interval might better approximate the duration of infectiousness.
Collapse
Affiliation(s)
- Erin C. Stone
- Hubert Department of Global Health, Laney Graduate School, Emory University, Atlanta, GA, USA
- Prevention and Response Branch, Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Devon L. Okasako-Schmucker
- Prevention and Response Branch, Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Joanna Taliano
- Office of Science Quality and Library Services (OSQLS), Office of Science (OS), Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Melissa Schaefer
- Prevention and Response Branch, Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - David T. Kuhar
- Hubert Department of Global Health, Laney Graduate School, Emory University, Atlanta, GA, USA
| |
Collapse
|
2
|
Chakraborty S, Cheng BYL, Edwards DL, Gonzalez JC, Chiu DKC, Zheng H, Scallan C, Guo X, Tan GS, Coffey GP, Conley PB, Hume PS, Janssen WJ, Byers DE, Mudd PA, Taubenberger J, Memoli M, Davis MM, Chua KF, Diamond MS, Andreakos E, Khatri P, Wang TT. Sialylated IgG induces the transcription factor REST in alveolar macrophages to protect against lung inflammation and severe influenza disease. Immunity 2025; 58:182-196.e10. [PMID: 39541970 PMCID: PMC11735284 DOI: 10.1016/j.immuni.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/15/2024] [Accepted: 10/04/2024] [Indexed: 11/17/2024]
Abstract
While most respiratory viral infections resolve with little harm to the host, severe symptoms arise when infection triggers an aberrant inflammatory response that damages lung tissue. Host regulators of virally induced lung inflammation have not been well defined. Here, we show that enrichment for sialylated, but not asialylated immunoglobulin G (IgG), predicted mild influenza disease in humans and was broadly protective against heterologous influenza viruses in a murine challenge model. Mechanistic studies show that sialylated IgG mediated this protection by inducing the transcription factor repressor element-1 silencing transcription factor (REST), which repressed nuclear factor κB (NF-κB)-driven responses, preventing severe lung inflammation and protecting lung function during influenza infection. Therapeutic administration of a recombinant, sialylated Fc molecule in clinical development similarly activated REST and protected against severe influenza disease, demonstrating that this pathway could be clinically harnessed. Overall, induction of REST through sialylated IgG signaling is a strategy to limit inflammatory disease sequelae in infections caused by antigenically distinct influenza strains.
Collapse
Affiliation(s)
- Saborni Chakraborty
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bowie Yik-Ling Cheng
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Desmond L Edwards
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Gonzalez
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David Kung-Chun Chiu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hong Zheng
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Courtney Scallan
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinrong Guo
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gene S Tan
- J. Craig Venter Institute, La Jolla, San Diego, CA 92037, USA; Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA 92037, USA
| | - Greg P Coffey
- Nuvig Therapeutics Inc., Redwood City, CA 94061, USA
| | | | - Patrick S Hume
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA
| | - William J Janssen
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Derek E Byers
- Department of Medicine, Division of Pulmonology and Critical Care Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Philip A Mudd
- Department of Emergency Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jeffery Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Matthew Memoli
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Mark M Davis
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; HHMI, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Katrin F Chua
- Department of Medicine, Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, CA 94305, USA; Geriatric Research, Education, and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Purvesh Khatri
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Center for Biomedical Informatics Research, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taia T Wang
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
3
|
Shetty N, Shephard MJ, Rockey NC, Macenczak H, Traenkner J, Danzy S, Vargas-Maldonado N, Arts PJ, Le Sage V, Anderson EJ, Lyon GM, Fitts EC, Gulick DA, Mehta AK, El-Chami MF, Kraft CS, Wigginton KR, Lowen AC, Marr LC, Rouphael NG, Lakdawala SS. Influenza virus infection and aerosol shedding kinetics in a controlled human infection model. J Virol 2024; 98:e0161224. [PMID: 39589151 DOI: 10.1128/jvi.01612-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/17/2024] [Indexed: 11/27/2024] Open
Abstract
Establishing effective mitigation strategies to reduce the spread of influenza virus requires an improved understanding of the mechanisms of transmission. We evaluated the use of a controlled human infection model using an H3N2 seasonal influenza virus to study critical aspects of transmission, including symptom progression and the dynamics of virus shedding. Eight volunteers were challenged with influenza A/Perth/16/2009 (H3N2) virus between July and September 2022 at Emory University Hospital. Viral shedding in the nasopharynx, saliva, stool, urine, and respiratory aerosols was monitored over the quarantine period, and symptoms were tracked until day 15. In addition, environmental swabs were collected from participant rooms to examine fomite contamination, and participant sera were collected to assess seroconversion by hemagglutination inhibition or microneutralization assays. Among the eight participants, influenza virus infection was confirmed in six (75%). Infectious virus or viral RNA was found in multiple physiological compartments, fecal samples, aerosol particles, and on surfaces in the immediate environment. Illness was moderate, with upper respiratory symptoms dominating. In participants with the highest viral loads, antibody titers rose by day 15 post-inoculation, while in participants with low or undetectable viral loads, there was little or no increase in functional antibody titers. These data demonstrate the safety and utility of the human infection model to study features critical to influenza virus transmission dynamics in a controlled manner and will inform the design of future challenge studies focused on modeling and limiting transmission.CLINICAL TRIALSThis study is registered with ClinicalTrials.gov as NCT05332899. IMPORTANCE We use a controlled human infection model to assess respiratory and aerosol shedding kinetics to expand our knowledge of influenza infection dynamics and help inform future studies aimed at understanding human-to-human transmission.
Collapse
Affiliation(s)
- Nishit Shetty
- Department of Civil and Environmental Engineering, Virginia Tech, Blacksburg, Virginia, USA
- Department of Civil, Environmental, and Architectural Engineering, University of Kansas, Lawrence, Kansas, USA
| | - Meredith J Shephard
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Nicole C Rockey
- Department of Civil and Environmental Engineering, Duke University, Durham, North Carolina, USA
| | | | | | - Shamika Danzy
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Nahara Vargas-Maldonado
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Peter J Arts
- Department of Civil and Environmental Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Valerie Le Sage
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Evan J Anderson
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - G Marshall Lyon
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Eric Charles Fitts
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Dalia A Gulick
- Department of Civil, Environmental, and Architectural Engineering, University of Kansas, Lawrence, Kansas, USA
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Aneesh K Mehta
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mikhael F El-Chami
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Colleen S Kraft
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Krista R Wigginton
- Department of Civil and Environmental Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Anice C Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Linsey C Marr
- Department of Civil and Environmental Engineering, Virginia Tech, Blacksburg, Virginia, USA
| | | | - Seema S Lakdawala
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Creppy JR, Delache B, Lemaitre J, Horvat B, Vecellio L, Ducancel F. Administration of airborne pathogens in non-human primates. Inhal Toxicol 2024; 36:475-500. [PMID: 39388247 DOI: 10.1080/08958378.2024.2412685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE Airborne pathogen scan penetrate in human respiratory tract and can cause illness. The use of animal models to predict aerosol deposition and study respiratory disease pathophysiology is therefore important for research and a prerequisite to test and study the mechanism of action of treatment. NHPs are relevant animal species for inhalation studies because of their similarities with humans in terms of anatomical structure, respiratory parameters and immune system. MATERIALS AND METHODS The aim of this review is to provide an overview of the state of the art of pathogen aerosol studies performed in non-human primates (NHPs). Herein, we present and discuss the deposition of aerosolized bacteria and viruses. In this review, we present important advantages of using NHPs as model for inhalation studies. RESULTS We demonstrate that deposition in the respiratory tract is not only a function of aerosol size but also the technique of administration influences the biological activity and site of aerosol deposition. Finally, we observe an influence of a region of pathogen deposition in the respiratory tract on the development of the pathophysiological effect in NHPs. CONCLUSION The wide range of methods used for the delivery of pathogento NHP respiratory airways is associated with varying doses and deposition profiles in the airways.
Collapse
Affiliation(s)
- Justina R Creppy
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
- Centre d'Étude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France
| | - Benoit Delache
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Julien Lemaitre
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Branka Horvat
- CIRI, Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, Université de Lyon, Lyon, France
| | - Laurent Vecellio
- Centre d'Étude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France
| | - Frédéric Ducancel
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| |
Collapse
|
5
|
Winchester S, Castellarnau A, Jabbar K, Nadir M, Ranasinghe K, Masramon X, Kinghorn GR, John I, Paull JRA. Astodrimer Sodium Nasal Spray versus Placebo in Non-Hospitalised Patients with COVID-19: A Randomised, Double-Blinded, Placebo-Controlled Trial. Pharmaceutics 2024; 16:1173. [PMID: 39339209 PMCID: PMC11435287 DOI: 10.3390/pharmaceutics16091173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND/OBJECTIVES Dendrimer-based astodrimer sodium nasal spray was assessed for its ability to reduce SARS-CoV-2 load in outpatients with COVID-19, which remains a severe illness for vulnerable groups. METHODS This was a randomised, double-blind, placebo-controlled clinical investigation evaluating the efficacy of astodrimer nasal spray in reducing SARS-CoV-2 viral burden in the nasopharynx of outpatients with COVID-19. Non-hospitalised adults with SARS-CoV-2 infection were randomised 1:1 to astodrimer or placebo four times daily from Day 1 to Day 7. Nasopharyngeal swabs for SARS-CoV-2 load determination were self-obtained daily from Day 1 to Day 8. The primary endpoint was an area under the curve of SARS-CoV-2 RNA copies/mL through Day 8 (vAUCd1-8). The primary analysis population was the modified intent-to-treat population (mITT: all randomised participants exposed to the study treatment who had at least one post-baseline viral load determination). Safety analyses included all randomised participants exposed to the study treatment. STUDY REGISTRATION ISRCTN70449927; Results: 231 participants were recruited between 9 January and 20 September 2023. The safety population comprised 109 and 113 participants randomised to astodrimer and placebo, respectively, with 96 and 101 participants in the mITT. Astodrimer sodium nasal spray reduced the SARS-CoV-2 burden (vAUCd1-8) vs. placebo in non-hospitalised COVID-19 patients aged 16 years and over (-1.2 log10 copies/mL × Day). The reduction in SARS-CoV-2 load was statistically significant in those aged 45 years and older (-3.7, p = 0.017) and the effect increased in older age groups, including in those aged 65 years and older (-7.3, p = 0.005). Astodrimer sodium nasal spray increased the rate of viral clearance and helped alleviate some COVID-19 symptoms, especially loss of sense of smell. Overall, 31 participants (14%) had ≥1 adverse event (AE). Four AEs were deemed possibly related to treatment. Most AEs were of mild severity and occurred at similar rates in both treatment arms. CONCLUSIONS Astodrimer nasal spray reduces viral burden and accelerates viral clearance, especially in older populations, and is well tolerated.
Collapse
Affiliation(s)
- Stephen Winchester
- Frimley Health National Health Service Foundation Trust, Camberley GU16 7UJ, UK
| | | | - Kashif Jabbar
- Ashford and St. Peter's Hospitals National Health Service Foundation Trust, Chertsey KT16 0PZ, UK
| | - Meera Nadir
- Ashford and St. Peter's Hospitals National Health Service Foundation Trust, Chertsey KT16 0PZ, UK
| | - Kapila Ranasinghe
- Ashford and St. Peter's Hospitals National Health Service Foundation Trust, Chertsey KT16 0PZ, UK
| | - Xavier Masramon
- Servicio de Asesoría a la Investigación y Logística (SAIL), 08027 Barcelona, Spain
| | - George R Kinghorn
- Royal Hallamshire Hospital, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Isaac John
- Ashford and St. Peter's Hospitals National Health Service Foundation Trust, Chertsey KT16 0PZ, UK
- Royal Holloway, University of London, Egham TW20 0EX, UK
| | | |
Collapse
|
6
|
Asplin P, Mancy R, Finnie T, Cumming F, Keeling MJ, Hill EM. Symptom propagation in respiratory pathogens of public health concern: a review of the evidence. J R Soc Interface 2024; 21:20240009. [PMID: 39045688 PMCID: PMC11267474 DOI: 10.1098/rsif.2024.0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/28/2024] [Indexed: 07/25/2024] Open
Abstract
Symptom propagation occurs when the symptom set an individual experiences is correlated with the symptom set of the individual who infected them. Symptom propagation may dramatically affect epidemiological outcomes, potentially causing clusters of severe disease. Conversely, it could result in chains of mild infection, generating widespread immunity with minimal cost to public health. Despite accumulating evidence that symptom propagation occurs for many respiratory pathogens, the underlying mechanisms are not well understood. Here, we conducted a scoping literature review for 14 respiratory pathogens to ascertain the extent of evidence for symptom propagation by two mechanisms: dose-severity relationships and route-severity relationships. We identify considerable heterogeneity between pathogens in the relative importance of the two mechanisms, highlighting the importance of pathogen-specific investigations. For almost all pathogens, including influenza and SARS-CoV-2, we found support for at least one of the two mechanisms. For some pathogens, including influenza, we found convincing evidence that both mechanisms contribute to symptom propagation. Furthermore, infectious disease models traditionally do not include symptom propagation. We summarize the present state of modelling advancements to address the methodological gap. We then investigate a simplified disease outbreak scenario, finding that under strong symptom propagation, isolating mildly infected individuals can have negative epidemiological implications.
Collapse
Affiliation(s)
- Phoebe Asplin
- EPSRC & MRC Centre for Doctoral Training in Mathematics for Real-World Systems, University of Warwick, Coventry, UK
- Mathematics Institute, University of Warwick, Coventry, UK
- The Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, University of Warwick, Coventry, UK
| | - Rebecca Mancy
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
- MRC/CSO Social and Public Health Sciences Unit, University of Glasgow, Glasgow, UK
| | - Thomas Finnie
- Data, Analytics and Surveillance, UK Health Security Agency, London, UK
| | - Fergus Cumming
- Foreign, Commonwealth and Development Office, London, UK
| | - Matt J. Keeling
- Mathematics Institute, University of Warwick, Coventry, UK
- The Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, University of Warwick, Coventry, UK
- School of Life Sciences, University of Glasgow, Glasgow, UK
| | - Edward M. Hill
- Mathematics Institute, University of Warwick, Coventry, UK
- The Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, University of Warwick, Coventry, UK
| |
Collapse
|
7
|
Jackson S, Marshall JL, Mawer A, Lopez-Ramon R, Harris SA, Satti I, Hughes E, Preston-Jones H, Cabrera Puig I, Longet S, Tipton T, Laidlaw S, Doherty RP, Morrison H, Mitchell R, Tanner R, Ateere A, Stylianou E, Wu MS, Fredsgaard-Jones TPW, Breuer J, Rapeport G, Ferreira VM, Gleeson F, Pollard AJ, Carroll M, Catchpole A, Chiu C, McShane H. Safety, tolerability, viral kinetics, and immune correlates of protection in healthy, seropositive UK adults inoculated with SARS-CoV-2: a single-centre, open-label, phase 1 controlled human infection study. THE LANCET. MICROBE 2024; 5:655-668. [PMID: 38703782 PMCID: PMC7616636 DOI: 10.1016/s2666-5247(24)00025-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND A SARS-CoV-2 controlled human infection model (CHIM) has been successfully established in seronegative individuals using a dose of 1×101 50% tissue culture infectious dose (TCID50) pre-alpha SARS-CoV-2 virus. Given the increasing prevalence of seropositivity to SARS-CoV-2, a CHIM that could be used for vaccine development will need to induce infection in those with pre-existing immunity. Our aim was to find a dose of pre-alpha SARS-CoV-2 virus that induced infection in previously infected individuals. METHODS Healthy, UK volunteers aged 18-30 years, with proven (quantitative RT-PCR or lateral flow antigen test) previous SARS-CoV-2 infection (with or without vaccination) were inoculated intranasally in a stepwise dose escalation CHIM with either 1×101, 1×102, 1×10³, 1×104, or 1×105 TCID50 SARS-CoV-2/human/GBR/484861/2020, the same virus used in the seronegative CHIM. Post-inoculation, volunteers were quarantined in functionally negative pressure rooms (Oxford, UK) for 14 days and until 12-hourly combined oropharyngeal-nasal swabs were negative for viable virus by focus-forming assay. Outpatient follow-up continued for 12 months post-enrolment, with additional visits for those who developed community-acquired SARS-CoV-2 infection. The primary objective was to identify a safe, well tolerated dose that induced infection (defined as two consecutive SARS-CoV-2 positive PCRs starting 24 h after inoculation) in 50% of seropositive volunteers. This study is registered with ClinicalTrials.gov (NCT04864548); enrolment and follow-up to 12 months post-enrolment are complete. FINDINGS Recruitment commenced on May 6, 2021, with the last volunteer enrolled into the dose escalation cohort on Nov 24, 2022. 36 volunteers were enrolled, with four to eight volunteers inoculated in each dosing group from 1×101 to 1×105 TCID50 SARS-CoV-2. All volunteers have completed quarantine, with follow-up to 12 months complete. Despite dose escalation to 1×105 TCID50, we were unable to induce sustained infection in any volunteers. Five (14%) of 36 volunteers were considered to have transient infection, based on the kinetic of their PCR-positive swabs. Transiently infected volunteers had significantly lower baseline mucosal and systemic SARS-CoV-2-specific antibody titres and significantly lower peripheral IFNγ responses against a CD8+ T-cell SARS-CoV-2 peptide pool than uninfected volunteers. 14 (39%) of 36 volunteers subsequently developed breakthrough infection with the omicron variant after discharge from quarantine. Most adverse events reported by volunteers in quarantine were mild, with fatigue (16 [44%]) and stuffy nose (16 [44%]) being the most common. There were no serious adverse events. INTERPRETATION Our study demonstrates potent protective immunity induced by homologous vaccination and homologous or heterologous previous SARS-CoV-2 infection. The community breakthrough infections seen with the omicron variant supports the use of newer variants to establish a model with sufficient rate of infection for use in vaccine and therapeutic development. FUNDING Wellcome Trust and Department for Health and Social Care.
Collapse
Affiliation(s)
- Susan Jackson
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Julia L Marshall
- Department of Paediatrics, University of Oxford, Oxford, UK; The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Andrew Mawer
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | | | - Iman Satti
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Eileen Hughes
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | | | - Stephanie Longet
- The Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tom Tipton
- The Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Stephen Laidlaw
- The Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Hazel Morrison
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Rachel Tanner
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Alberta Ateere
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Meng-San Wu
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Judith Breuer
- Institute of Child Health, University College London, London, UK
| | - Garth Rapeport
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Fergus Gleeson
- Oxford Radiology Research Unit, University of Oxford, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Miles Carroll
- The Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Helen McShane
- Department of Paediatrics, University of Oxford, Oxford, UK.
| |
Collapse
|
8
|
Pérez-Díez A, Liu X, Calderon S, Bennett A, Lisco A, Kellog A, Galindo F, Memoli MJ, Rocco JM, Epling BP, Laidlaw E, Sneller MC, Manion M, Wortmann GW, Poon R, Kumar P, Sereti I. Prevalence of anti-lymphocyte IgM autoantibodies driving complement activation in COVID-19 patients. Front Immunol 2024; 15:1352330. [PMID: 38694513 PMCID: PMC11061367 DOI: 10.3389/fimmu.2024.1352330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/27/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction COVID-19 patients can develop autoantibodies against a variety of secreted and membrane proteins, including some expressed on lymphocytes. However, it is unclear what proportion of patients might develop anti-lymphocyte antibodies (ALAb) and what functional relevance they might have. Methods We evaluated the presence and lytic function of ALAb in the sera of a cohort of 85 COVID-19 patients (68 unvaccinated and 17 vaccinated) assigned to mild (N=63), or moderate/severe disease (N=22) groups. Thirty-seven patients were followed-up after recovery. We also analyzed in vivo complement deposition on COVID-19 patients' lymphocytes and examined its correlation with lymphocyte numbers during acute disease. Results Compared with healthy donors (HD), patients had an increased prevalence of IgM ALAb, which was significantly higher in moderate/severe disease patients and persisted after recovery. Sera from IgM ALAb+ patients exhibited complement-dependent cytotoxicity (CDC) against HD lymphocytes. Complement protein C3b deposition on patients' CD4 T cells was inversely correlated with CD4 T cell numbers. This correlation was stronger in moderate/severe disease patients. Discussion IgM ALAb and complement activation against lymphocytes may contribute to the acute lymphopenia observed in COVID-19 patients.
Collapse
Affiliation(s)
- Ainhoa Pérez-Díez
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Xiangdong Liu
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Stephanie Calderon
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Ashlynn Bennett
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Andrea Lisco
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Anela Kellog
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Frances Galindo
- Division of Clinical Research, NIAID, NIH, Bethesda, MD, United States
| | - Matthew J. Memoli
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Joseph M. Rocco
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Brian P. Epling
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Elizabeth Laidlaw
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Mike C. Sneller
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Maura Manion
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Glenn W. Wortmann
- Section of Infectious Diseases, MedStar Washington Hospital Center, Washington, DC, United States
| | - Rita Poon
- Division of Hospital Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Princy Kumar
- Division of Infectious Diseases and Tropical Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Irini Sereti
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| |
Collapse
|
9
|
Bean R, Giurgea LT, Han A, Czajkowski L, Cervantes-Medina A, Gouzoulis M, Mateja A, Hunsberger S, Reed S, Athota R, Baus HA, Kash JC, Park J, Taubenberger JK, Memoli MJ. Mucosal correlates of protection after influenza viral challenge of vaccinated and unvaccinated healthy volunteers. mBio 2024; 15:e0237223. [PMID: 38193710 PMCID: PMC10865821 DOI: 10.1128/mbio.02372-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
The induction of systemic antibody titers against hemagglutinin has long been the main focus of influenza vaccination strategies, but mucosal immunity has also been shown to play a key role in the protection against respiratory viruses. By vaccinating and challenging healthy volunteers, we demonstrated that inactivated influenza vaccine (IIV) modestly reduced the rate of influenza while predominantly boosting serum antibody titers against hemagglutinin (HA) and HA stalk, a consequence of the low neuraminidase (NA) content of IIV and the intramuscular route of administration. The viral challenge induced nasal and serum responses against both HA and NA. Correlations between mucosal IgA and serum IgG against specific antigens were low, whether before or after challenge, suggesting a compartmentalization of immune responses. Even so, volunteers who developed viral shedding for multiple days had lower baseline titers across both systemic and mucosal compartments as compared to those with no shedding or a single day of shedding. Regression analysis showed that pre-challenge HA inhibition titers were the most consistent correlate of protection across clinical outcomes combining shedding and symptoms, with NA inhibition titers and HA IgG levels only predicting the duration of shedding. Despite the inclusion of data from multiple binding and functional antibody assays against HA and NA performed on both serum and nasal samples, multivariate models were unable to account for the variability in outcomes, emphasizing our imperfect understanding of immune correlates in influenza and the importance of refining models with assessments of innate and cellular immune responses.IMPORTANCEThe devastating potential of influenza has been well known for over 100 years. Despite the development of vaccines since the middle of the 20th century, influenza continues to be responsible for substantial global morbidity and mortality. To develop next-generation vaccines with enhanced effectiveness, we must synthesize our understanding of the complex immune mechanisms culminating in protection. Our study outlines the differences in immune responses to influenza vaccine and influenza infection, identifying potential gaps in vaccine-induced immunity, particularly at the level of the nasal mucosa. Furthermore, this research underscores the need to refine our imperfect models while recognizing potential pitfalls in past and future attempts to identify and measure correlates of protection.
Collapse
Affiliation(s)
- Rachel Bean
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Luca T. Giurgea
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Alison Han
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lindsay Czajkowski
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Adriana Cervantes-Medina
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Monica Gouzoulis
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Allyson Mateja
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Sally Hunsberger
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Susan Reed
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Rani Athota
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Holly Ann Baus
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - John C. Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jaekeun Park
- Department of Veterinary Medicine, VA-MD College of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Jeffery K. Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthew J. Memoli
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Oxford JS, Catchpole A, Mann A, Bell A, Noulin N, Gill D, Oxford JR, Gilbert A, Balasingam S. A Brief History of Human Challenge Studies (1900-2021) Emphasising the Virology, Regulatory and Ethical Requirements, Raison D'etre, Ethnography, Selection of Volunteers and Unit Design. Curr Top Microbiol Immunol 2024; 445:1-32. [PMID: 35704095 DOI: 10.1007/82_2022_253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Venetian quarantine 400 years ago was an important public health measure. Since 1900 this has been refined to include "challenge" or deliberate infection with pathogens be they viruses, bacteria, or parasites. Our focus is virology and ranges from the early experiments in Cuba with Yellow Fever Virus to the most widespread pathogen of our current times, COVID-19. The latter has so far caused over four million deaths worldwide and 190 million cases of the disease. Quarantine and challenge were also used to investigate the Spanish Influenza of 1918 which caused over 100 million deaths. We consider here the merits of the approach, that is the speeding up of knowledge in a practical sense leading to the more rapid licensing of vaccines and antimicrobials. At the core of quarantine and challenge initiatives is the design of the unit to allow safe confinement of the pathogen and protection of the staff. Most important though is the safety of volunteers. We can see now, as in 1900, that members of our society are prepared and willing to engage in these experiments for the public good. Our ethnology study, where the investigator observed the experiment from within the quarantine, gave us the first indication of changing attitudes amongst volunteers whilst in quarantine. These quarantine experiments, referred to as challenge studies, human infection studies, or "controlled human infection models" involve thousands of clinical samples taken over two to three weeks and can provide a wealth of immunological and molecular data on the infection itself and could allow the discovery of new targets for vaccines and therapeutics. The Yellow Fever studies from 121 years ago gave the impetus for development of a successful vaccine still used today whilst also uncovering the nature of the Yellow Fever agent, namely that it was a virus. We outline how carefully these experiments are approached and the necessity to have high quality units with self-contained air-flow along with extensive personal protective equipment for nursing and medical staff. Most important is the employment of highly trained scientific, medical and nursing staff. We face a future of emerging pathogens driven by the increasing global population, deforestation, climate change, antibiotic resistance and increased global travel. These emerging pathogens may be pathogens we currently are not aware of or have not caused outbreaks historically but could also be mutated forms of known pathogens including viruses such as influenza (H7N9, H5N1 etc.) and coronaviruses. This calls for challenge studies to be part of future pandemic preparedness as an additional tool to assist with the rapid development of broad-spectrum antimicrobials, immunomodulators and new vaccines.
Collapse
Affiliation(s)
- J S Oxford
- Blizzard Institute of Cell and Molecular Science, Queen Mary University of London, London, E1 2AT, UK
| | | | | | | | | | - D Gill
- Blizzard Institute of Cell and Molecular Science, Queen Mary University of London, London, E1 2AT, UK
| | - J R Oxford
- Inveresk Medical Practice, Edinburgh, E21 7BP, UK
| | | | | |
Collapse
|
11
|
Clarkson KA, Porter CK, Talaat KR, Kapulu MC, Chen WH, Frenck RW, Bourgeois AL, Kaminski RW, Martin LB. Shigella-Controlled Human Infection Models: Current and Future Perspectives. Curr Top Microbiol Immunol 2024; 445:257-313. [PMID: 35616717 PMCID: PMC7616482 DOI: 10.1007/82_2021_248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Shigella-controlled human infection models (CHIMs) are an invaluable tool utilized by the vaccine community to combat one of the leading global causes of infectious diarrhea, which affects infants, children and adults regardless of socioeconomic status. The impact of shigellosis disproportionately affects children in low- and middle-income countries (LMICs) resulting in cognitive and physical stunting, perpetuating a cycle that must be halted. Shigella-CHIMs not only facilitate the early evaluation of enteric countermeasures and up-selection of the most promising products but also provide insight into mechanisms of infection and immunity that are not possible utilizing animal models or in vitro systems. The greater understanding of shigellosis obtained in CHIMs builds and empowers the development of new generation solutions to global health issues which are unattainable in the conventional laboratory and clinical settings. Therefore, refining, mining and expansion of safe and reproducible infection models hold the potential to create effective means to end diarrheal disease and associated co-morbidities associated with Shigella infection.
Collapse
Affiliation(s)
- Kristen A Clarkson
- Department of Diarrheal Disease Research, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Chad K Porter
- Enteric Disease Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Kawsar R Talaat
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, 624 North Broadway Street Hampton House, Baltimore, MD, 21205, USA
| | - Melissa C Kapulu
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi County Hospital, Off Bofa Road, Kilifi, 80108, Kenya
| | - Wilbur H Chen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Robert W Frenck
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - A Louis Bourgeois
- PATH Center for Vaccine Innovation and Access, 455 Massachusetts Avenue NW, Washington, DC, 20001, USA
| | - Robert W Kaminski
- Department of Diarrheal Disease Research, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Laura B Martin
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100, Siena, Italy.
| |
Collapse
|
12
|
ElSherif M, Halperin SA. Benefits of Combining Molecular Biology and Controlled Human Infection Model Methodologies in Advancing Vaccine Development. J Mol Biol 2023; 435:168322. [PMID: 37866477 DOI: 10.1016/j.jmb.2023.168322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Infectious diseases continue to account for a significant portion of global deaths despite the use of vaccines for several centuries. Immunization programs around the world are a testament to the great success of multiple vaccines, yet there are still diseases without vaccines and others that require safer more effective ones. Addressing uncontrolled and emerging disease threats is restrained by the limitations and bottlenecks encountered with traditional vaccine development paradigms. Recent advances in modern molecular biology technologies have enhanced the interrogation of host pathogen interaction and deciphered complex pathways, thereby uncovering the myriad interplay of biological events that generate immune protection against foreign agents. Consequent to insights into the immune system, modern biology has been instrumental in the development and production of next generation 21st century vaccines. As these biological tools, commonly and collectively referred to as 'omics, became readily available, there has been a renewed consideration of Controlled Human Infection Models (CHIMs). Successful and reproducible CHIMs can complement modern molecular biology for the study of infectious diseases and development of effective vaccines in a regulated process that mitigates risk, cost, and time, with capacity to discern immune correlates of protection.
Collapse
Affiliation(s)
- May ElSherif
- Canadian Center for Vaccinology, IWK Health, Nova Scotia Health, and Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Scott A Halperin
- Canadian Center for Vaccinology, IWK Health, Nova Scotia Health, and Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
13
|
Styles CT, Zhou J, Flight KE, Brown JC, Lewis C, Wang X, Vanden Oever M, Peacock TP, Wang Z, Millns R, O'Neill JS, Borodavka A, Grove J, Barclay WS, Tregoning JS, Edgar RS. Propylene glycol inactivates respiratory viruses and prevents airborne transmission. EMBO Mol Med 2023; 15:e17932. [PMID: 37970627 PMCID: PMC10701621 DOI: 10.15252/emmm.202317932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 11/17/2023] Open
Abstract
Viruses are vulnerable as they transmit between hosts, and we aimed to exploit this critical window. We found that the ubiquitous, safe, inexpensive and biodegradable small molecule propylene glycol (PG) has robust virucidal activity. Propylene glycol rapidly inactivates a broad range of viruses including influenza A, SARS-CoV-2 and rotavirus and reduces disease burden in mice when administered intranasally at concentrations commonly found in nasal sprays. Most critically, vaporised PG efficiently abolishes influenza A virus and SARS-CoV-2 infectivity within airborne droplets, potently preventing infection at levels well below those tolerated by mammals. We present PG vapour as a first-in-class non-toxic airborne virucide that can prevent transmission of existing and emergent viral pathogens, with clear and immediate implications for public health.
Collapse
Affiliation(s)
| | - Jie Zhou
- Department of Infectious DiseaseImperial College LondonLondonUK
| | - Katie E Flight
- Department of Infectious DiseaseImperial College LondonLondonUK
- Present address:
University College LondonLondonUK
| | | | - Charlotte Lewis
- MRC‐University of Glasgow Centre for Virus ResearchGlasgowUK
| | - Xinyu Wang
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - Michael Vanden Oever
- Department of Infectious DiseaseImperial College LondonLondonUK
- Present address:
Life Edit TherapeuticsMorrisvilleNCUSA
| | | | - Ziyin Wang
- Department of Infectious DiseaseImperial College LondonLondonUK
| | - Rosie Millns
- Department of Infectious DiseaseImperial College LondonLondonUK
| | | | | | - Joe Grove
- MRC‐University of Glasgow Centre for Virus ResearchGlasgowUK
| | - Wendy S Barclay
- Department of Infectious DiseaseImperial College LondonLondonUK
| | | | - Rachel S Edgar
- Department of Infectious DiseaseImperial College LondonLondonUK
| |
Collapse
|
14
|
Abo YN, Jamrozik E, McCarthy JS, Roestenberg M, Steer AC, Osowicki J. Strategic and scientific contributions of human challenge trials for vaccine development: facts versus fantasy. THE LANCET. INFECTIOUS DISEASES 2023; 23:e533-e546. [PMID: 37573871 DOI: 10.1016/s1473-3099(23)00294-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 08/15/2023]
Abstract
The unprecedented speed of delivery of SARS-CoV-2 pandemic vaccines has redefined the limits for all vaccine development. Beyond the aspirational 100-day timeline for tomorrow's hypothetical pandemic vaccines, there is a sense of optimism that development of other high priority vaccines can be accelerated. Early in the COVID-19 pandemic, an intense and polarised academic and public discourse arose concerning the role of human challenge trials for vaccine development. A case was made for human challenge trials as a powerful tool to establish early proof-of-concept of vaccine efficacy in humans, inform vaccine down selection, and address crucial knowledge gaps regarding transmission, pathogenesis, and immune protection. We review the track record of human challenge trials contributing to the development of vaccines for 19 different pathogens and discuss relevant limitations, barriers, and pitfalls. This Review also highlights opportunities for efforts to broaden the scope and boost the effects of human challenge trials, to accelerate all vaccine development.
Collapse
Affiliation(s)
- Yara-Natalie Abo
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia; Infectious Diseases Unit, Department of General Medicine, Royal Children's Hospital Melbourne, Parkville, VIC, Australia.
| | - Euzebiusz Jamrozik
- Ethox and Pandemic Sciences Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK; Monash-WHO Collaborating Centre for Bioethics, Monash University, Melbourne, VIC, Australia
| | - James S McCarthy
- Department of Infectious Diseases, The University of Melbourne, Parkville, VIC, Australia; Victorian Infectious Diseases Services, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Meta Roestenberg
- Controlled Human Infections Center, Leiden University Medical Center, Leiden, Netherlands
| | - Andrew C Steer
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia; Infectious Diseases Unit, Department of General Medicine, Royal Children's Hospital Melbourne, Parkville, VIC, Australia
| | - Joshua Osowicki
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia; Infectious Diseases Unit, Department of General Medicine, Royal Children's Hospital Melbourne, Parkville, VIC, Australia
| |
Collapse
|
15
|
Le Sage V, Lowen AC, Lakdawala SS. Block the Spread: Barriers to Transmission of Influenza Viruses. Annu Rev Virol 2023; 10:347-370. [PMID: 37308086 DOI: 10.1146/annurev-virology-111821-115447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Respiratory viruses, such as influenza viruses, cause significant morbidity and mortality worldwide through seasonal epidemics and sporadic pandemics. Influenza viruses transmit through multiple modes including contact (either direct or through a contaminated surface) and inhalation of expelled aerosols. Successful human to human transmission requires an infected donor who expels virus into the environment, a susceptible recipient, and persistence of the expelled virus within the environment. The relative efficiency of each mode can be altered by viral features, environmental parameters, donor and recipient host characteristics, and viral persistence. Interventions to mitigate transmission of influenza viruses can target any of these factors. In this review, we discuss many aspects of influenza virus transmission, including the systems to study it, as well as the impact of natural barriers and various nonpharmaceutical and pharmaceutical interventions.
Collapse
Affiliation(s)
- Valerie Le Sage
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anice C Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA;
| | - Seema S Lakdawala
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA;
| |
Collapse
|
16
|
Anderson TL, Nande A, Merenstein C, Raynor B, Oommen A, Kelly BJ, Levy MZ, Hill AL. Quantifying individual-level heterogeneity in infectiousness and susceptibility through household studies. Epidemics 2023; 44:100710. [PMID: 37556994 PMCID: PMC10594662 DOI: 10.1016/j.epidem.2023.100710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/17/2023] [Accepted: 07/18/2023] [Indexed: 08/11/2023] Open
Abstract
The spread of SARS-CoV-2, like that of many other pathogens, is governed by heterogeneity. "Superspreading," or "over-dispersion," is an important factor in transmission, yet it is hard to quantify. Estimates from contact tracing data are prone to potential biases due to the increased likelihood of detecting large clusters of cases, and may reflect variation in contact behavior more than biological heterogeneity. In contrast, the average number of secondary infections per contact is routinely estimated from household surveys, and these studies can minimize biases by testing all members of a household. However, the models used to analyze household transmission data typically assume that infectiousness and susceptibility are the same for all individuals or vary only with predetermined traits such as age. Here we develop and apply a combined forward simulation and inference method to quantify the degree of inter-individual variation in both infectiousness and susceptibility from observations of the distribution of infections in household surveys. First, analyzing simulated data, we show our method can reliably ascertain the presence, type, and amount of these heterogeneities given data from a sufficiently large sample of households. We then analyze a collection of household studies of COVID-19 from diverse settings around the world, and find strong evidence for large heterogeneity in both the infectiousness and susceptibility of individuals. Our results also provide a framework to improve the design of studies to evaluate household interventions in the presence of realistic heterogeneity between individuals.
Collapse
Affiliation(s)
- Thayer L Anderson
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, United States of America
| | - Anjalika Nande
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, United States of America
| | - Carter Merenstein
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Brinkley Raynor
- Department of Biostatistics, Epidemiology, & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Anisha Oommen
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, United States of America; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, United States of America
| | - Brendan J Kelly
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America; Department of Biostatistics, Epidemiology, & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America; Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Michael Z Levy
- Department of Biostatistics, Epidemiology, & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Alison L Hill
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, United States of America; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, United States of America.
| |
Collapse
|
17
|
Ortiz JR, Bernstein DI, Hoft DF, Woods CW, McClain MT, Frey SE, Brady RC, Bryant C, Wegel A, Frenck RW, Walter EB, Abate G, Williams SR, Atmar RL, Keitel WA, Rouphael N, Memoli MJ, Makhene MK, Roberts PC, Neuzil KM. A Multicenter, Controlled Human Infection Study of Influenza A(H1N1)pdm09 in Healthy Adults. J Infect Dis 2023; 228:287-298. [PMID: 36702771 PMCID: PMC10420403 DOI: 10.1093/infdis/jiad021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND We evaluated the associations between baseline influenza virus-specific hemagglutination inhibition (HAI) and microneutralization (MN) titers and subsequent symptomatic influenza virus infection in a controlled human infection study. METHODS We inoculated unvaccinated healthy adults aged 18-49 years with an influenza A/California/04/2009/H1N1pdm-like virus (NCT04044352). We collected serial safety labs, serum for HAI and MN, and nasopharyngeal swabs for reverse-transcription polymerase chain reaction (RT-PCR) testing. Analyses used the putative seroprotective titer of ≥40 for HAI and MN. The primary clinical outcome was mild-to-moderate influenza disease (MMID), defined as ≥1 postchallenge positive qualitative RT-PCR test with a qualifying symptom/clinical finding. RESULTS Of 76 participants given influenza virus challenge, 54 (71.1%) experienced MMID. Clinical illness was generally very mild. MMID attack rates among participants with baseline titers ≥40 by HAI and MN were 64.9% and 67.9%, respectively, while MMID attack rates among participants with baseline titers <40 by HAI and MN were 76.9% and 78.3%, respectively. The estimated odds of developing MMID decreased by 19% (odds ratio, 0.81 [95% confidence interval, .62-1.06]; P = .126) for every 2-fold increase in baseline HAI. There were no significant adverse events. CONCLUSIONS We achieved a 71.1% attack rate of MMID. High baseline HAI and MN were associated with protection from illness. Clinical Trials Registration. NCT04044352.
Collapse
Affiliation(s)
- Justin R Ortiz
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - David I Bernstein
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Ohio; Departments of
| | - Daniel F Hoft
- Internal Medicine and
- Molecular Microbiology and Immunology, Division of Infectious Diseases, Allergy and Immunology and Center for Vaccine Development, Saint Louis University School of Medicine, Missouri
| | - Christopher W Woods
- Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina
| | - Micah T McClain
- Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina
| | | | - Rebecca C Brady
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Ohio; Departments of
| | - Christopher Bryant
- Vaccine and Infectious Disease Therapeutic Research Unit, The Emmes Company, Rockville, Maryland
| | - Ashley Wegel
- Vaccine and Infectious Disease Therapeutic Research Unit, The Emmes Company, Rockville, Maryland
| | - Robert W Frenck
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Ohio; Departments of
| | - Emmanuel B Walter
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina
| | | | - Sarah R Williams
- Division of Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore
| | - Robert L Atmar
- Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Wendy A Keitel
- Departments of Molecular Virology & Microbiology and Medicine, Baylor College of Medicine, Houston, Texas
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | | | - Mamodikoe K Makhene
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Paul C Roberts
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
18
|
Zsichla L, Müller V. Risk Factors of Severe COVID-19: A Review of Host, Viral and Environmental Factors. Viruses 2023; 15:175. [PMID: 36680215 PMCID: PMC9863423 DOI: 10.3390/v15010175] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
The clinical course and outcome of COVID-19 are highly variable, ranging from asymptomatic infections to severe disease and death. Understanding the risk factors of severe COVID-19 is relevant both in the clinical setting and at the epidemiological level. Here, we provide an overview of host, viral and environmental factors that have been shown or (in some cases) hypothesized to be associated with severe clinical outcomes. The factors considered in detail include the age and frailty, genetic polymorphisms, biological sex (and pregnancy), co- and superinfections, non-communicable comorbidities, immunological history, microbiota, and lifestyle of the patient; viral genetic variation and infecting dose; socioeconomic factors; and air pollution. For each category, we compile (sometimes conflicting) evidence for the association of the factor with COVID-19 outcomes (including the strength of the effect) and outline possible action mechanisms. We also discuss the complex interactions between the various risk factors.
Collapse
Affiliation(s)
- Levente Zsichla
- Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
- National Laboratory for Health Security, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Viktor Müller
- Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
- National Laboratory for Health Security, Eötvös Loránd University, 1117 Budapest, Hungary
| |
Collapse
|
19
|
Jay C, Ratcliff J, Turtle L, Goulder P, Klenerman P. Exposed seronegative: Cellular immune responses to SARS-CoV-2 in the absence of seroconversion. Front Immunol 2023; 14:1092910. [PMID: 36776841 PMCID: PMC9909393 DOI: 10.3389/fimmu.2023.1092910] [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: 11/08/2022] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
The factors determining whether infection will occur following exposure to SARS-CoV-2 remain elusive. Certain SARS-CoV-2-exposed individuals mount a specific T-cell response but fail to seroconvert, representing a population that may provide further clarity on the nature of infection susceptibility and correlates of protection against SARS-CoV-2. Exposed seronegative individuals have been reported in patients exposed to the blood-borne pathogens Human Immunodeficiency virus and Hepatitis C virus and the sexually transmitted viruses Hepatitis B virus and Herpes Simplex virus. By comparing the quality of seronegative T-cell responses to SARS-CoV-2 with seronegative cellular immunity to these highly divergent viruses, common patterns emerge that offer insights on the role of cellular immunity against infection. For both SARS-CoV-2 and Hepatitis C, T-cell responses in exposed seronegatives are consistently higher than in unexposed individuals, but lower than in infected, seropositive patients. Durability of T-cell responses to Hepatitis C is dependent upon repeated exposure to antigen - single exposures do not generate long-lived memory T-cells. Finally, exposure to SARS-CoV-2 induces varying degrees of immune activation, suggesting that exposed seronegative individuals represent points on a spectrum rather than a discrete group. Together, these findings paint a complex landscape of the nature of infection but provide clues as to what may be protective early on in SARS-CoV-2 disease course. Further research on this phenomenon, particularly through cohort studies, is warranted.
Collapse
Affiliation(s)
- Cecilia Jay
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jeremy Ratcliff
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Lance Turtle
- National Institute for Health and Care Research Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, United Kingdom
| | - Philip Goulder
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Anderson TL, Nande A, Merenstein C, Raynor B, Oommen A, Kelly BJ, Levy MZ, Hill AL. Quantifying individual-level heterogeneity in infectiousness and susceptibility through household studies. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.12.02.22281853. [PMID: 36523404 PMCID: PMC9753792 DOI: 10.1101/2022.12.02.22281853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The spread of SARS-CoV-2, like that of many other pathogens, is governed by heterogeneity. "Superspreading," or "over-dispersion," is an important factor in transmission, yet it is hard to quantify. Estimates from contact tracing data are prone to potential biases due to the increased likelihood of detecting large clusters of cases, and may reflect variation in contact behavior more than biological heterogeneity. In contrast, the average number of secondary infections per contact is routinely estimated from household surveys, and these studies can minimize biases by testing all members of a household. However, the models used to analyze household transmission data typically assume that infectiousness and susceptibility are the same for all individuals or vary only with predetermined traits such as age. Here we develop and apply a combined forward simulation and inference method to quantify the degree of inter-individual variation in both infectiousness and susceptibility from observations of the distribution of infections in household surveys. First, analyzing simulated data, we show our method can reliably ascertain the presence, type, and amount of these heterogeneities with data from a sufficiently large sample of households. We then analyze a collection of household studies of COVID-19 from diverse settings around the world, and find strong evidence for large heterogeneity in both the infectiousness and susceptibility of individuals. Our results also provide a framework to improve the design of studies to evaluate household interventions in the presence of realistic heterogeneity between individuals.
Collapse
Affiliation(s)
- Thayer L Anderson
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218
| | - Anjalika Nande
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218
| | - Carter Merenstein
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Brinkley Raynor
- Department of Biostatistics, Epidemiology, & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Anisha Oommen
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Brendan J Kelly
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Michael Z Levy
- Department of Biostatistics, Epidemiology, & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Alison L Hill
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218
| |
Collapse
|
21
|
Joseph J, Baby HM, Zhao S, Li X, Cheung K, Swain K, Agus E, Ranganathan S, Gao J, Luo JN, Joshi N. Role of bioaerosol in virus transmission and material-based countermeasures. EXPLORATION (BEIJING, CHINA) 2022; 2:20210038. [PMID: 37324804 PMCID: PMC10190935 DOI: 10.1002/exp.20210038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/15/2022] [Indexed: 06/17/2023]
Abstract
Respiratory pathogens transmit primarily through particles such as droplets and aerosols. Although often overlooked, the resuspension of settled droplets is also a key facilitator of disease transmission. In this review, we discuss the three main mechanisms of aerosol generation: direct generation such as coughing and sneezing, indirect generation such as medical procedures, and resuspension of settled droplets and aerosols. The size of particles and environmental factors influence their airborne lifetime and ability to cause infection. Specifically, humidity and temperature are key factors controlling the evaporation of suspended droplets, consequently affecting the duration in which particles remain airborne. We also suggest material-based approaches for effective prevention of disease transmission. These approaches include electrostatically charged virucidal agents and surface coatings, which have been shown to be highly effective in deactivating and reducing resuspension of pathogen-laden aerosols.
Collapse
Affiliation(s)
- John Joseph
- Center for Nanomedicine, Department of AnesthesiologyPerioperative and Pain Medicine, Brigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Helna Mary Baby
- Center for Nanomedicine, Department of AnesthesiologyPerioperative and Pain Medicine, Brigham and Women's HospitalBostonMassachusettsUSA
| | - Spencer Zhao
- Center for Nanomedicine, Department of AnesthesiologyPerioperative and Pain Medicine, Brigham and Women's HospitalBostonMassachusettsUSA
| | - Xiang‐Ling Li
- Center for Nanomedicine, Department of AnesthesiologyPerioperative and Pain Medicine, Brigham and Women's HospitalBostonMassachusettsUSA
| | - Krisco‐Cheuk Cheung
- Center for Nanomedicine, Department of AnesthesiologyPerioperative and Pain Medicine, Brigham and Women's HospitalBostonMassachusettsUSA
| | - Kabir Swain
- Center for Nanomedicine, Department of AnesthesiologyPerioperative and Pain Medicine, Brigham and Women's HospitalBostonMassachusettsUSA
| | - Eli Agus
- Center for Nanomedicine, Department of AnesthesiologyPerioperative and Pain Medicine, Brigham and Women's HospitalBostonMassachusettsUSA
| | - Sruthi Ranganathan
- Center for Nanomedicine, Department of AnesthesiologyPerioperative and Pain Medicine, Brigham and Women's HospitalBostonMassachusettsUSA
| | - Jingjing Gao
- Center for Nanomedicine, Department of AnesthesiologyPerioperative and Pain Medicine, Brigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - James N Luo
- Harvard Medical SchoolBostonMassachusettsUSA
- Department of SurgeryBrigham and Women's HospitalBostonMassachusettsUSA
| | - Nitin Joshi
- Center for Nanomedicine, Department of AnesthesiologyPerioperative and Pain Medicine, Brigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
22
|
Choy RKM, Bourgeois AL, Ockenhouse CF, Walker RI, Sheets RL, Flores J. Controlled Human Infection Models To Accelerate Vaccine Development. Clin Microbiol Rev 2022; 35:e0000821. [PMID: 35862754 PMCID: PMC9491212 DOI: 10.1128/cmr.00008-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The timelines for developing vaccines against infectious diseases are lengthy, and often vaccines that reach the stage of large phase 3 field trials fail to provide the desired level of protective efficacy. The application of controlled human challenge models of infection and disease at the appropriate stages of development could accelerate development of candidate vaccines and, in fact, has done so successfully in some limited cases. Human challenge models could potentially be used to gather critical information on pathogenesis, inform strain selection for vaccines, explore cross-protective immunity, identify immune correlates of protection and mechanisms of protection induced by infection or evoked by candidate vaccines, guide decisions on appropriate trial endpoints, and evaluate vaccine efficacy. We prepared this report to motivate fellow scientists to exploit the potential capacity of controlled human challenge experiments to advance vaccine development. In this review, we considered available challenge models for 17 infectious diseases in the context of the public health importance of each disease, the diversity and pathogenesis of the causative organisms, the vaccine candidates under development, and each model's capacity to evaluate them and identify correlates of protective immunity. Our broad assessment indicated that human challenge models have not yet reached their full potential to support the development of vaccines against infectious diseases. On the basis of our review, however, we believe that describing an ideal challenge model is possible, as is further developing existing and future challenge models.
Collapse
Affiliation(s)
- Robert K. M. Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | - A. Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Richard I. Walker
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Jorge Flores
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| |
Collapse
|
23
|
Antonio EC, Meireles MR, Bragatte MADS, Vieira GF. Viral immunogenic footprints conferring T cell cross-protection to SARS-CoV-2 and its variants. Front Immunol 2022; 13:931372. [PMID: 35967415 PMCID: PMC9366040 DOI: 10.3389/fimmu.2022.931372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
COVID-19 brought scenes from sci-fi movies into real life. Infected individuals include asymptomatic cases to severe disease leading to death, suggesting the involvement of the genetic constitution of populations and pathogens contributing to differential individuals' outcomes. To investigate shared immunogenic features between SARS-CoV-2 targets and other coronaviruses, we modeled their peptides in 3D structures of HLA-A*02:01 (pMHC), comparing their molecular surfaces These structures were also compared with a panel of epitopes from unrelated viruses, looking for potential triggers conferring cross-protection in uninfected individuals. As expected, SARS-CoV 1 and 2 peptides share molecular and physicochemical features, providing an explanation for the verified experimental immunogenicity among them. Surprisingly, even discordant sequences from human coronaviruses 229E, OC43 and epitopes from unrelated viruses involved in endemic human infections exhibit similar fingerprints of immunogenicity with SARS-CoV-2 peptides. The same approach indicates a conserved CD8+ T cell recognition between Wuhan SARS-CoV-2 sequences and altered peptides from Variants of Concern. Examination of structural data over epitope sequence analysis here could explain how previous infections may produce a heterologous immunity response in a global scale against emergent diseases such as Covid-19, mitigating its full lethal potential, and paves the way for the development of wide spectrum vaccine development.
Collapse
Affiliation(s)
- Eduardo Cheuiche Antonio
- Post Graduation Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mariana Rost Meireles
- Post Graduation Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Gustavo Fioravanti Vieira
- Post Graduation Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Post Graduation Program in Health and Human Development, Universidade La Salle Canoas, Canoas, Brazil
| |
Collapse
|
24
|
Kim JW, Kwak SG, Lee H, Kim SK, Choe JY, Park SH. Baseline use of hydroxychloroquine or immunosuppressive drugs and the risk of coronavirus disease 2019. Korean J Intern Med 2022; 37:673-680. [PMID: 33706472 PMCID: PMC9082427 DOI: 10.3904/kjim.2020.633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/26/2020] [Accepted: 01/18/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIMS The preventive role of hydroxychloroquine (HCQ) on coronavirus disease 2019 (COVID-19) remains unclear. The aim of this study was to examine the effects of HCQ and other immunosuppressive drugs on the incidence of COVID-19. METHODS The data were collected from the South Korea National Health Insurance Sharing-COVID-19 database. All individuals who underwent nasopharyngeal and oropharyngeal swab tests for COVID-19 from January 2020 to May 2020 are included. The association between COVID-19 risk and HCQ use was examined in a propensity score-matched population. Factors associated with COVID-19 were identified using multiple logistic regression analysis. RESULTS Total 8,070 patients with COVID-19 and 121,050 negative controls were included from the database. Among all participants, 381 were HCQ users. In a propensity score-matched population, the incidence of COVID-19 was 7.1% in HCQ users and 6.8% in non-users. The odds ratio (OR) for HCQ use was 1.05 with a 95% confidence interval (CI) of 0.58 to 1.89. Among the subpopulation of patients with rheumatoid arthritis (RA), 33 were diagnosed with COVID-19 and 478 were not. Use of HCQ, glucocorticoids, or other immunosuppressive drugs was not associated with COVID-19 risk, whereas abatacept use was. Chronic lung disease was an independent risk factor for COVID-19 diagnosis in patients with RA (adjusted OR, 6.07; 95% CI, 1.10 to 33.59). CONCLUSION The risk of COVID-19 did not differ between HCQ users and non-users. Glucocorticoids, conventional disease-modifying antirheumatic drugs (DMARDs), and biological DMARDs other than abatacept did not increase the risk of COVID-19.
Collapse
Affiliation(s)
- Ji-Won Kim
- Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Sang Gyu Kwak
- Department of Medical Statistics, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Hwajeong Lee
- Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Seong-Kyu Kim
- Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Jung-Yoon Choe
- Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Sung-Hoon Park
- Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
| |
Collapse
|
25
|
Balasingam S, Meillon S, Chui C, Mann A, La C, Weller CL, King DF, Smith E. Human infection studies: Key considerations for challenge agent development and production. Wellcome Open Res 2022; 7:140. [PMID: 35505774 PMCID: PMC9034172 DOI: 10.12688/wellcomeopenres.17869.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2022] [Indexed: 12/03/2022] Open
Abstract
Human infection (or challenge) studies involve the intentional administration of a pathogen (challenge agent) to volunteers. The selection, isolation, development and production of the challenge agent is one of the first steps in developing a challenge study and critical for minimising the risk to volunteers. Regulatory oversight for this production differs globally. Manufacturing agents within a Good Manufacturing Practice (GMP) facility reduces the risk of the manufacturing process by including processes such as confirming the identity of the challenge agent and ascertaining that it's pure and free from impurities. However, in some cases it's not possible or feasible to manufacture to GMP standards, for example where the challenge agent requires an intermediate vector for growth. There is lack of clear guidance on what the minimum requirements for high-quality safe manufacture outside of GMP facilities should be and here we describe the development of a considerations document for the selection and production of challenge agents to meet this need.
Collapse
Affiliation(s)
| | - Sarah Meillon
- Infectious Diseases
- Prevention, Wellcome Trust, UK, London, N1 2BE, UK
| | | | - Alex Mann
- Global Health Innovative Technology Fund, Tokyo 106-0032, Japan
| | - Carine La
- Global Health Innovative Technology Fund, Tokyo 106-0032, Japan
| | | | - Deborah F. King
- Infectious Diseases
- Prevention, Wellcome Trust, UK, London, N1 2BE, UK
| | - Emma Smith
- HIC-Vac, Imperial College London, National Heart & Lung Institute (NHLI), London, W2 1PG, UK
| |
Collapse
|
26
|
Motallebi S, Cheung RCY, Mohit B, Shahabi S, Alishahi Tabriz A, Moattari S. Modeling COVID-19 Mortality Across 44 Countries: Face Covering May Reduce Deaths. Am J Prev Med 2022; 62:483-491. [PMID: 35305777 PMCID: PMC8580811 DOI: 10.1016/j.amepre.2021.09.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/07/2021] [Accepted: 09/13/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Despite ongoing efforts to vaccinate communities against COVID-19, the necessity of face mask use in controlling the pandemic remains subject to debate. Several studies have investigated face masks and COVID-19, covering smaller and less diverse populations than this study's sample. This study examines a hypothesized association of face-covering mandates with COVID-19 mortality decline across 44 countries in 2 continents. METHODS In a retrospective cohort study, changes in COVID-19‒related daily mortality rate per million population from February 15 to May 31, 2020 were compared between 27 countries with and 17 countries without face mask mandates in nearly 1 billion (911,446,220 total) people. Longitudinal mixed effect modeling was applied and adjusted for over 10 relevant demographic, social, clinical, and time-dependent confounders. RESULTS Average COVID-19 mortality per million was 288.54 in countries without face mask policies and 48.40 in countries with face mask policies. In no mask countries, adjusted average daily increase was 0.1553 - 0.0017 X (days since the first case) log deaths per million, compared with 0.0900 - 0.0009 X (days since the first case) log deaths per million in the countries with a mandate. A total of 60 days into the pandemic, countries without face mask mandates had an average daily increase of 0.0533 deaths per million, compared with the average daily increase of 0.0360 deaths per million for countries with face mask mandates. CONCLUSIONS This study's significant results show that face mask mandates were associated with lower COVID-19 deaths rates than the rates in countries without mandates. These findings support the use of face masks to prevent excess COVID-19 deaths and should be advised during airborne disease epidemics.
Collapse
Affiliation(s)
- Sahar Motallebi
- Department of Social Medicine and Global Health, Lund University, Lund, Sweden
| | - Rex C Y Cheung
- Department of Decision Sciences, San Francisco State University, San Francisco, California
| | - Babak Mohit
- Sleep Disorders Center, University of Maryland Medical Center, Baltimore, Maryland.
| | - Shahram Shahabi
- Southwest College of Naturopathic Medicine & Health Sciences, Tempe, Arizona
| | - Amir Alishahi Tabriz
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | - Syamak Moattari
- Health Sciences Department, Worcester State University, Worcester, Massachusetts
| |
Collapse
|
27
|
Affiliation(s)
- Kathryn M Edwards
- Division of Infectious Diseases, Department of Pediatrics, Monroe Carell Jr Children's Hospital at Vanderbilt, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
28
|
Horie NC, Schmid K, Silva BFFD. COVID-19 among workers assisting homeless and socially vulnerable people. EINSTEIN-SAO PAULO 2022; 20:eAO6237. [PMID: 35293526 PMCID: PMC8909120 DOI: 10.31744/einstein_journal/2022ao6237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/18/2021] [Indexed: 11/17/2022] Open
Abstract
Objective To describe the profile of professionals assisting homeless and socially vulnerable populations tested for COVID-19, and to determine potential associations with exposure at the workplace, on the way to work, or at home, among infected professionals. To describe disease symptoms and progression and to investigate potential associations with age, sex and exposure at the workplace, on the way to work, or at home. Methods A retrospective analysis of data of 173 workers employed by Serviço Franciscano de Solidariedade tested for SARS-CoV-2. Between May 20 and June 2, 2020, professionals and volunteers were tested for anti-SARS-CoV-2 IgG and IgM antibodies, by means of qualitative rapid chromatographic immunoassay in whole blood. A questionnaire was used to collect data on demographic characteristics and working conditions, history and date of onset of symptoms and risk factors. Quantitative variables were expressed as mean and standard deviation, or median, maximum, and minimum values. Data normality was investigated using the Kolmogorov-Smirnov test. Results A total of 46 (26.6%) participants had positive serologic tests. Of participants with negative serologic test results, 109 (85.8%) were asymptomatic. History of symptoms was the most significant independent factor associated with positive serology. Serologic test results and symptoms differed significantly according to housing (p=0.045) and working (p<0.001) conditions. More than half of participants (52.4%) living in shared households tested positive, compared to 23% of participants living in family households. Participants working remotely from home did not test positive. In seropositive participants, onset of symptoms was associated with workplace exposure and shared housing conditions. Conclusion History of symptoms was associated with positive serology for COVID-19. Shared housing conditions tended to be associated with higher risk of infection. Onset of symptoms was associated with higher levels of workplace exposure and shared housing conditions in seropositive participants.
Collapse
|
29
|
Acer Ö, Bahçe YG, Özüdoğru O. Association of viral load with age, gender, disease severity and death in SARS-CoV-2 variants. J Med Virol 2022; 94:3063-3069. [PMID: 35212012 DOI: 10.1002/jmv.27677] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/02/2022] [Accepted: 02/21/2022] [Indexed: 11/12/2022]
Abstract
In the current study, the relationship between viral load, demographic characteristics, and disease information in 1007(48.5%) patients with Delta variant (B.1.617.2), and 1070 (51.5%) patients with Alpha variant (B1.1.7) mutations was investigated. We found that there was a significant difference in viral load between patients who died from the Alpha variant and those who were discharged (p<0.05). Nevertheless, no significant difference was observed in patients with Delta variant. The viral load in patients who died from the Alpha variant was significantly higher than those who were discharged (p<0.05). The viral load was found to be higher in females in patients with Delta variant, whereas it was very close in males and females in patients with Alpha variant (p>0.05). No significant difference was detected between the cycle threshold values (Ct) and disease severity. In terms of the mean Ct values, statistical differences were observed in patients with Delta and Alpha variant. The Alpha variant was found to have a higher viral load than the Delta variant. Furthermore, Delta variant was found to be higher in the 40-year-old and under-age group than in the Alpha variant, whereas the Alpha variant was higher in the 40-year-old and older group. While the rate of moderate and severe patients in Alpha variant was found to be higher, the rate of mild survivors was found to be higher in Delta variant. In conclusion, The increase in vaccination prior to the appearance of the Delta variant in our region may have influenced the viral load and clinical status of the patients. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ömer Acer
- Siirt University, Medical Faculty, Department of Medical Microbiology, 56100, Siirt, Turkey
| | - Yasemin Genç Bahçe
- Siirt Training and Research Hospital, Microbiology Laboratory, 56100, Siirt, Turkey
| | - Osman Özüdoğru
- Siirt University, Medical Faculty, Department of Internal Medicine, 56100, Siirt, Turkey
| |
Collapse
|
30
|
Abstract
During infection, the rates of pathogen replication, death, and migration affect disease progression, dissemination, transmission, and resistance evolution. Here, we follow the population dynamics of Vibrio cholerae in a mouse model by labeling individual bacteria with one of >500 unique, fitness-neutral genomic tags. Using the changes in tag frequencies and CFU numbers, we inform a mathematical model that describes the within-host spatiotemporal bacterial dynamics. This allows us to disentangle growth, death, forward, and retrograde migration rates continuously during infection. Our model has robust predictive power across various experimental setups. The population dynamics of V. cholerae shows substantial spatiotemporal heterogeneity in replication, death, and migration. Importantly, we find that the niche available to V. cholerae in the host increases with inoculum size, suggesting cooperative effects during infection. Therefore, it is not enough to consider just the likelihood of exposure (50% infectious dose) but rather the magnitude of exposure to predict outbreaks. IMPORTANCE Determining the rates of bacterial migration, replication, and death during infection is important for understanding how infections progress. Separately measuring these rates is often difficult in systems where multiple processes happen simultaneously. Here, we use next-generation sequencing to measure V. cholerae migration, replication, death, and niche size along the mouse gastrointestinal tract. We show that the small intestine of the mouse is a heterogeneous environment, and the population dynamic characteristics change substantially between adjacent gut sections. Our approach also allows us to characterize the effect of inoculum size on these processes. We find that the niche size in mice increases with the infectious dose, hinting at cooperative effects in larger inocula. The dose-response relationship between inoculum size and final pathogen burden is important for the infected individual and is thought to influence the progression of V. cholerae epidemics.
Collapse
|
31
|
London V, Blitz MJ, Solmonovich R, Silver M, Minkoff H. Temporal Trend in SARS-CoV-2 Symptoms in Pregnant Women. Am J Perinatol 2021. [PMID: 34918329 DOI: 10.1055/s-0041-1739490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE The objective of this study was to examine temporal trends in the clinical presentation of patients diagnosed with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in pregnancy. STUDY DESIGN This is a retrospective cohort study of pregnant women who were universally screened for SARS-CoV-2 and tested positive. This multi-center study of admissions to labor and delivery units in New York City and Long Island included all SARS-CoV-2-infected pregnant women admitted to labor and delivery units between April 10th and June 4th 2020. Six Northwell Health hospitals and Maimonides Medical Center were included in the study. The main measures of the study included patient reports of COVID-19 symptoms: fever, cough, chest pain, shortness of breath, nausea, vomiting, and intensive care unit (ICU) admissions. The main outcome measure was the percentage of all infected women who reported any of the above symptoms. RESULTS In total, 427 infected pregnant women were included in the study. There was a statistically significant decline in the percentage of patients presenting with any symptoms over the course of the study. In addition, disease severity, symptoms of fever, cough, and chest pain/shortness of breath also significantly declined over time, and no ICU admissions were noted after the third week of April. CONCLUSIONS There was a temporal shift away from symptomatic presentation in pregnant women diagnosed with SARS-CoV-2 over the course of the first months of the epidemic in New York. Further studies are necessary to elucidate the cause of this change in presentation among pregnant women, to determine whether this trend is also observed in other patient populations. KEY POINTS · Retrospective cohort review of 427 SARS-CoV-2-infected pregnant women admitted to labor and delivery units.. · A significant decline in the percentage of patients presenting with symptoms over time was noted.. · Further studies are necessary to elucidate the cause of this change in presentation.. · Theories for the noted trend: viral evolution, decreased viral inoculums, and prolonged polymerase chain reaction positivity..
Collapse
Affiliation(s)
- Viktoriya London
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Maimonides Medical Center, Brooklyn, New York
| | - Matthew J Blitz
- Department of Obstetrics and Gynecology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York.,Division of Maternal-Fetal Medicine, Southside Hospital, Bay Shore, New York
| | - Rachel Solmonovich
- Department of Obstetrics and Gynecology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - Michael Silver
- Department of Obstetrics and Gynecology, SUNY Downstate Medical Center, Brooklyn, New York
| | - Howard Minkoff
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Maimonides Medical Center, Brooklyn, New York.,Department of Obstetrics and Gynecology, SUNY Downstate Medical Center, Brooklyn, New York
| |
Collapse
|
32
|
Immune-mediated attenuation of influenza illness after infection: opportunities and challenges. THE LANCET MICROBE 2021; 2:e715-e725. [DOI: 10.1016/s2666-5247(21)00180-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/01/2021] [Accepted: 07/01/2021] [Indexed: 01/04/2023] Open
|
33
|
Chapman PR, Giacomin P, Loukas A, McCarthy JS. Experimental human hookworm infection: a narrative historical review. PLoS Negl Trop Dis 2021; 15:e0009908. [PMID: 34882670 PMCID: PMC8659326 DOI: 10.1371/journal.pntd.0009908] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In 1896, a serendipitous laboratory accident led to the understanding that hookworms propagate infection by penetrating skin, a theory that was then confirmed with the first experimental human infection, reported in 1901. Experimental human infections undertaken in the 20th century enabled understanding of the natural history of infection and the immune response. More recently, experimental hookworm infection has been performed to investigate the immunomodulatory potential of hookworm infection and for the evaluation of hookworm vaccines and chemotherapeutic interventions. Experimental human hookworm infection has been proven to be safe, with no deaths observed in over 500 participants (although early reports predate systematic adverse event reporting) and no serious adverse events described in over 200 participants enrolled in contemporary clinical trials. While experimental human hookworm infection holds significant promise, as both a challenge model for testing anti-hookworm therapies and for treating various diseases of modernity, there are many challenges that present. These challenges include preparation and storage of larvae, which has not significantly changed since Harada and Mori first described their coproculture method in 1955. In vitro methods of hookworm larval culture, storage, and the development of meaningful potency or release assays are required. Surrogate markers of intestinal infection intensity are required because faecal egg counts or hookworm faecal DNA intensity lack the fidelity required for exploration of hookworm infection as a vaccine/drug testing platform or as a regulated therapy.
Collapse
Affiliation(s)
- Paul R. Chapman
- Clinical Tropical Medicine, QIMR Berghofer Medical Research Institute, Herston, Australia
- Infectious Diseases Unit, Royal Brisbane and Women’s Hospital, Herston, Australia
| | - Paul Giacomin
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - James S. McCarthy
- Clinical Tropical Medicine, QIMR Berghofer Medical Research Institute, Herston, Australia
- Infectious Diseases Unit, Royal Brisbane and Women’s Hospital, Herston, Australia
| |
Collapse
|
34
|
Blom KB, Åsberg A, Sjaastad I, Kalleberg KT, Søraas A, Midtvedt K, Birkeland JA. Kidney Transplant Recipient Behavior During the Early COVID-19 Pandemic: A National Survey Study in Norway. Kidney Med 2021; 4:100389. [PMID: 34805967 PMCID: PMC8596763 DOI: 10.1016/j.xkme.2021.09.006] [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: 12/02/2022] Open
Abstract
Rationale & Objective Studies published from countries with a high prevalence of COVID-19 have found increased incidence and a more severe disease course of coronavirus disease 2019 (COVID-19) in kidney transplant recipients than in the general population. We investigated how the first wave of the COVID-19 pandemic affected the everyday life of kidney transplant recipients in a country with a low infection burden. Study Design Prospective case-control study. Setting & Participants All adult kidney transplant recipients in Norway with a functioning graft and listed in the public phone registry (n = 3,060) and a group of randomly recruited individuals >18 years from the general population (n = 20,000) were invited to participate in the study by an SMS text message. In parallel, all kidney transplant recipients in Norway were invited to measure severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) IgG from mid-June to October 2020. Predictors The participants were asked to fill out a questionnaire focused on everyday life, travel history, exposure to known COVID-19 cases, and demographics. Analytical Approach Groups were compared with independent tests using 2-sided 0.05 significance levels. Results A total of 1,007 kidney transplant recipients and 4,409 controls answered the questionnaire. The kidney transplant recipients reported being more concerned about SARS-CoV-2 infection (27%) than the control group (7%; P value < 0.001); ie, they behaved more carefully in their everyday life (not going to the grocery store, 5.9% vs 0.9%, P < 0.001; keeping at least 1 meter distance, 16.6% vs 5.8%, P < 0.001). Of the kidney transplant responders, 81% had a SARS-CoV-2 IgG taken; all were negative. Limitations Mortality data is not reliable because of the low number of SARS-CoV-2 infected kidney transplant recipients in Norway. The relatively low questionnaire response rate for kidney transplant recipients is not optimal. Conclusions The questionnaire shows that kidney transplant recipients have behaved more carefully compared with the general population with less social interaction and a very high degree of adherence to governmental advice.
Collapse
Affiliation(s)
- Kjersti B Blom
- Department of Nephrology, Oslo University Hospital, Ullevål, Oslo, Norway.,Institute for Experimental Medical Research, Oslo University Hospital, Ullevål, Oslo, Norway and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Anders Åsberg
- Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital, Ullevål, Oslo, Norway and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway.,Department of Cardiology, Oslo University Hospital, Ullevål, Oslo, Norway
| | | | - Arne Søraas
- Department of Microbiology, Oslo University Hospital, Case Comprehensive Cancer Center, Oslo, Norway
| | - Karsten Midtvedt
- Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Jon A Birkeland
- Department of Nephrology, Oslo University Hospital, Ullevål, Oslo, Norway
| |
Collapse
|
35
|
Kollepara PK, Siegenfeld AF, Taleb NN, Bar-Yam Y. Unmasking the mask studies: why the effectiveness of surgical masks in preventing respiratory infections has been underestimated. J Travel Med 2021; 28:taab144. [PMID: 34490465 PMCID: PMC8499874 DOI: 10.1093/jtm/taab144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 11/14/2022]
Abstract
BACKGROUND Pre-pandemic empirical studies have produced mixed statistical results on the effectiveness of masks against respiratory viruses, leading to confusion that may have contributed to organizations such as the WHO and CDC initially not recommending that the general public wear masks during the coronavirus disease 2019 pandemic. METHODS A threshold-based dose-response curve framework is used to analyse the effects of interventions on infection probabilities for both single and repeated exposure events. Empirical studies on mask effectiveness are evaluated with a statistical power analysis that includes the effect of adherence to mask usage protocols. RESULTS When the adherence to mask usage guidelines is taken into account, the empirical evidence indicates that masks prevent disease transmission: all studies we analysed that did not find surgical masks to be effective were under-powered to such an extent that even if masks were 100% effective, the studies in question would still have been unlikely to find a statistically significant effect. We also provide a framework for understanding the effect of masks on the probability of infection for single and repeated exposures. The framework demonstrates that masks can have a disproportionately large protective effect and that more frequently wearing a mask provides super-linearly compounding protection. CONCLUSIONS This work shows (1) that both theoretical and empirical evidence is consistent with masks protecting against respiratory infections and (2) that non-linear effects and statistical considerations regarding the percentage of exposures for which masks are worn must be taken into account when designing empirical studies and interpreting their results.
Collapse
Affiliation(s)
- Pratyush K Kollepara
- New England Complex Systems Institute, Cambridge, MA, USA
- Department of Physics, BITS Pilani K K Birla Goa Campus, Goa, India
| | - Alexander F Siegenfeld
- New England Complex Systems Institute, Cambridge, MA, USA
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Yaneer Bar-Yam
- New England Complex Systems Institute, Cambridge, MA, USA
| |
Collapse
|
36
|
Spinelli MA, Glidden DV, Gennatas ED, Bielecki M, Beyrer C, Rutherford G, Chambers H, Goosby E, Gandhi M. Importance of non-pharmaceutical interventions in lowering the viral inoculum to reduce susceptibility to infection by SARS-CoV-2 and potentially disease severity. THE LANCET. INFECTIOUS DISEASES 2021; 21:e296-e301. [PMID: 33631099 PMCID: PMC7906703 DOI: 10.1016/s1473-3099(20)30982-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 01/01/2023]
Abstract
Adherence to non-pharmaceutical interventions to prevent the transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been highly variable across settings, particularly in the USA. In this Personal View, we review data supporting the importance of the viral inoculum (the dose of viral particles from an infected source over time) in increasing the probability of infection in respiratory, gastrointestinal, and sexually transmitted viral infections in humans. We also review the available evidence linking the relationship of the viral inoculum to disease severity. Non-pharmaceutical interventions might reduce the susceptibility to SARS-CoV-2 infection by reducing the viral inoculum when there is exposure to an infectious source. Data from physical sciences research suggest that masks protect the wearer by filtering virus from external sources, and others by reducing expulsion of virus by the wearer. Social distancing, handwashing, and improved ventilation also reduce the exposure amount of viral particles from an infectious source. Maintaining and increasing non-pharmaceutical interventions can help to quell SARS-CoV-2 as we enter the second year of the pandemic. Finally, we argue that even as safe and effective vaccines are being rolled out, non-pharmaceutical interventions will continue to play an essential role in suppressing SARS-CoV-2 transmission until equitable and widespread vaccine administration has been completed.
Collapse
Affiliation(s)
- Matthew A Spinelli
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - David V Glidden
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Efstathios D Gennatas
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Michel Bielecki
- Swiss Armed Forces, Medical Services, Ittigen, Switzerland; Travel Clinic, Institute for Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Chris Beyrer
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - George Rutherford
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Henry Chambers
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Eric Goosby
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Monica Gandhi
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
37
|
Burman J. The use of respirators and its impact on the COVID-19 pandemic in Europe between 1 June and 14 October 2020. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2021; 62:E625-E627. [PMID: 34909489 PMCID: PMC8639117 DOI: 10.15167/2421-4248/jpmh2021.62.3.1909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/25/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Cases of COVID-19 infection have increased sharply in Europe since August 2020, and the WHO recommend the use of respirators in situations where keeping distance is not possible. The purpose of this study was to evaluate the impact of the use of respirators on infection cases and viral deaths in European countries. METHODS COVID-19 cases and related deaths in 29 countries relative to population were searched through the WHO database on 15.10.2020. Recent 14-day cases in relation to the population were retrieved from the European Center for Disease Prevention and Control's website, covering the period from 1 to 14 October 2020. Information related to the use of respirators was retrieved from the IHME database of the University of Washington at time point, 1.6.2020. RESULTS The proportion of people using of respirators at 1.6.2020 correlate negatively to the reported cases of disease (Rs = -0.528) and to deaths (Rs = -0.553). No significant correlation was found for recent cases or mortality. Countries with at least 60% respirator use did not differ from other countries. CONCLUSION Long-term use of respirators appears to reduce disease incidence and death in the population.
Collapse
Affiliation(s)
- Janne Burman
- University of Helsinki and Helsinki University Hospital, Finland
| |
Collapse
|
38
|
Viral and Bacterial Co-Infections in the Lungs: Dangerous Liaisons. Viruses 2021; 13:v13091725. [PMID: 34578306 PMCID: PMC8472850 DOI: 10.3390/v13091725] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/23/2022] Open
Abstract
Respiratory tract infections constitute a significant public health problem, with a therapeutic arsenal that remains relatively limited and that is threatened by the emergence of antiviral and/or antibiotic resistance. Viral–bacterial co-infections are very often associated with the severity of these respiratory infections and have been explored mainly in the context of bacterial superinfections following primary influenza infection. This review summarizes our current knowledge of the mechanisms underlying these co-infections between respiratory viruses (influenza viruses, RSV, and SARS-CoV-2) and bacteria, at both the physiological and immunological levels. This review also explores the importance of the microbiome and the pathological context in the evolution of these respiratory tract co-infections and presents the different in vitro and in vivo experimental models available. A better understanding of the complex functional interactions between viruses/bacteria and host cells will allow the development of new, specific, and more effective diagnostic and therapeutic approaches.
Collapse
|
39
|
Giurgea LT, Cervantes-Medina A, Walters KA, Scherler K, Han A, Czajkowski LM, Baus HA, Hunsberger S, Klein SL, Kash JC, Taubenberger JK, Memoli MJ. Sex Differences in Influenza: The Challenge Study Experience. J Infect Dis 2021; 225:715-722. [PMID: 34423369 DOI: 10.1093/infdis/jiab422] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/19/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Preclinical animal studies and retrospective human studies suggest that adult females have worse outcomes from influenza than males. Prospective studies in humans are missing. METHODS Data from 164 healthy volunteers who underwent Influenza A/California/04/2009/H1N1 challenge were compiled to compare differences between sexes. Baseline characteristics, including hormone levels, hemagglutination-inhibition (HAI) titers, neuraminidase-inhibition titers (NAI), and outcomes after challenge were compared. Linear and logistic regression models were built to determine significant predictor variables with respect to outcomes of interest. RESULTS Hemagglutination-inhibition (HAI) titers were similar between the sexes, but neuraminidase-inhibition titers (NAI) were higher in males than females at 4-weeks and 8-weeks post-challenge. Females were more likely to have symptoms (mean 0.96 vs 0.80, p=.003) and to have a higher number of symptoms (median 3 vs 4, p=.011) than males. Linear and logistic regression models showed that pre-challenge NAI titers, but not HAI titers or sex hormone levels, were predictive of all shedding and symptom outcomes of interest. CONCLUSIONS Females in our cohorts were more likely to be symptomatic and to have a higher number of symptoms than males. NAI titers predicted all outcomes of interest and may explain differential outcomes between the sexes.
Collapse
Affiliation(s)
- Luca T Giurgea
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Dr., Bethesda, MD 20892 USA
| | - Adriana Cervantes-Medina
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Dr., Bethesda, MD 20892 USA
| | | | | | - Alison Han
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Dr., Bethesda, MD 20892 USA
| | - Lindsay M Czajkowski
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Dr., Bethesda, MD 20892 USA
| | - Holly Ann Baus
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Dr., Bethesda, MD 20892 USA
| | - Sally Hunsberger
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20894 USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - John C Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Dr., Bethesda, MD 20892 USA
| | - Jeffery K Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Dr., Bethesda, MD 20892 USA
| | - Matthew J Memoli
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Dr., Bethesda, MD 20892 USA
| |
Collapse
|
40
|
Lin X, Lin F, Liang T, Ducatez MF, Zanin M, Wong SS. Antibody Responsiveness to Influenza: What Drives It? Viruses 2021; 13:v13071400. [PMID: 34372607 PMCID: PMC8310379 DOI: 10.3390/v13071400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/02/2021] [Accepted: 07/03/2021] [Indexed: 02/06/2023] Open
Abstract
The induction of a specific antibody response has long been accepted as a serological hallmark of recent infection or antigen exposure. Much of our understanding of the influenza antibody response has been derived from studying antibodies that target the hemagglutinin (HA) protein. However, growing evidence points to limitations associated with this approach. In this review, we aim to highlight the issue of antibody non-responsiveness after influenza virus infection and vaccination. We will then provide an overview of the major factors known to influence antibody responsiveness to influenza after infection and vaccination. We discuss the biological factors such as age, sex, influence of prior immunity, genetics, and some chronic infections that may affect the induction of influenza antibody responses. We also discuss the technical factors, such as assay choices, strain variations, and viral properties that may influence the sensitivity of the assays used to measure influenza antibodies. Understanding these factors will hopefully provide a more comprehensive picture of what influenza immunogenicity and protection means, which will be important in our effort to improve influenza vaccines.
Collapse
Affiliation(s)
- Xia Lin
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
| | - Fangmei Lin
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
| | - Tingting Liang
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
| | | | - Mark Zanin
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
- School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Sook-San Wong
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
- School of Public Health, The University of Hong Kong, Hong Kong, China
- Correspondence: ; Tel.: +86-178-2584-6078
| |
Collapse
|
41
|
Goyal A, Reeves DB, Thakkar N, Famulare M, Cardozo-Ojeda EF, Mayer BT, Schiffer JT. Slight reduction in SARS-CoV-2 exposure viral load due to masking results in a significant reduction in transmission with widespread implementation. Sci Rep 2021; 11:11838. [PMID: 34088959 PMCID: PMC8178300 DOI: 10.1038/s41598-021-91338-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 05/12/2021] [Indexed: 12/19/2022] Open
Abstract
Masks are a vital tool for limiting SARS-CoV-2 spread in the population. Here we utilize a mathematical model to assess the impact of masking on transmission within individual transmission pairs and at the population level. Our model quantitatively links mask efficacy to reductions in viral load and subsequent transmission risk. Our results reinforce that the use of masks by both a potential transmitter and exposed person substantially reduces the probability of successful transmission, even if masks only lower exposure viral load by ~ 50%. Slight increases in mask adherence and/or efficacy above current levels would reduce the effective reproductive number (Re) substantially below 1, particularly if implemented comprehensively in potential super-spreader environments. Our model predicts that moderately efficacious masks will also lower exposure viral load tenfold among people who get infected despite masking, potentially limiting infection severity. Because peak viral load tends to occur pre-symptomatically, we also identify that antiviral therapy targeting symptomatic individuals is unlikely to impact transmission risk. Instead, antiviral therapy would only lower Re if dosed as post-exposure prophylaxis and if given to ~ 50% of newly infected people within 3 days of an exposure. These results highlight the primacy of masking relative to other biomedical interventions under consideration for limiting the extent of the COVID-19 pandemic prior to widespread implementation of a vaccine. To confirm this prediction, we used a regression model of King County, Washington data and simulated the counterfactual scenario without mask wearing to estimate that in the absence of additional interventions, mask wearing decreased Re from 1.3-1.5 to ~ 1.0 between June and September 2020.
Collapse
Affiliation(s)
- Ashish Goyal
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Daniel B Reeves
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Niket Thakkar
- Institute for Disease Modeling, Global Health Division, Bill and Melinda Gates Foundation, Seattle, USA
| | - Mike Famulare
- Institute for Disease Modeling, Global Health Division, Bill and Melinda Gates Foundation, Seattle, USA
| | - E Fabián Cardozo-Ojeda
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Bryan T Mayer
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Joshua T Schiffer
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, USA.
- Department of Medicine, University of Washington, Seattle, USA.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.
| |
Collapse
|
42
|
Drossinos Y, Weber TP, Stilianakis NI. Droplets and aerosols: An artificial dichotomy in respiratory virus transmission. Health Sci Rep 2021; 4:e275. [PMID: 33977157 PMCID: PMC8103093 DOI: 10.1002/hsr2.275] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/08/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
In the medical literature, three mutually non-exclusive modes of pathogen transmission associated with respiratory droplets are usually identified: contact, droplet, and airborne (or aerosol) transmission. The demarcation between droplet and airborne transmission is often based on a cut-off droplet diameter, most commonly 5 μm. We argue here that the infectivity of a droplet, and consequently the transmissivity of the virus, as a function of droplet size is a continuum, depending on numerous factors (gravitational settling rate, transport, and dispersion in a turbulent air jet, viral load and viral shedding, virus inactivation) that cannot be adequately characterized by a single droplet diameter. We propose instead that droplet and aerosol transmission should be replaced by a unique airborne transmission mode, to be distinguished from contact transmission.
Collapse
Affiliation(s)
- Yannis Drossinos
- Joint Research CentreEuropean Commission, Joint Research Centre (JRC)IspraItaly
| | - Thomas P. Weber
- Joint Research CentreEuropean Commission, Directorate General for Health and Food SafetyBrusselsBelgium
| | - Nikolaos I. Stilianakis
- Joint Research CentreEuropean Commission, Joint Research Centre (JRC)IspraItaly
- Department of Biometry and EpidemiologyUniversity of Erlangen‐NurembergErlangenGermany
| |
Collapse
|
43
|
Cortese M, Sherman AC, Rouphael NG, Pulendran B. Systems Biological Analysis of Immune Response to Influenza Vaccination. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a038596. [PMID: 32152245 DOI: 10.1101/cshperspect.a038596] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The last decade has witnessed tremendous progress in immunology and vaccinology, owing to several scientific and technological breakthroughs. Systems vaccinology is a field that has emerged at the forefront of vaccine research and development and provides a unique way to probe immune responses to vaccination in humans. The goals of systems vaccinology are to use systems-based approaches to define signatures that can be used to predict vaccine immunogenicity and efficacy and to delineate the molecular mechanisms driving protective immunity. The application of systems biological approaches in influenza vaccination studies has enabled the discovery of early signatures that predict immunogenicity to vaccination and yielded novel mechanistic insights about vaccine-induced immunity. Here we review the contributions of systems vaccinology to influenza vaccine development and critically examine the potential of systems vaccinology toward enabling the development of a universal influenza vaccine that provides robust and durable immunity against diverse influenza viruses.
Collapse
Affiliation(s)
- Mario Cortese
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, California 94305, USA
| | - Amy C Sherman
- Hope Clinic of the Emory Vaccine Center, Decatur, Georgia 30030, USA
| | - Nadine G Rouphael
- Hope Clinic of the Emory Vaccine Center, Decatur, Georgia 30030, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, California 94305, USA.,Department of Pathology, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, California 94305, USA.,Department of Pathology, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
44
|
Falahi S, Abdoli A, Kenarkoohi A. Claims and reasons about mild COVID-19 in children. New Microbes New Infect 2021; 41:100864. [PMID: 33747533 PMCID: PMC7963516 DOI: 10.1016/j.nmni.2021.100864] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 02/28/2021] [Accepted: 03/10/2021] [Indexed: 12/22/2022] Open
Abstract
The elderly form the main risk group in the coronavirus disease 2019 (COVID-19) pandemic, and age is recognized as a major risk factor for the severity of infection and mortality of COVID-19. The severity of the infection in children is milder than in adults. Although the pathophysiology of COVID-19 is not fully understood, several possible factors and mechanisms have been suggested for the lower severity of infection in children.
Collapse
Affiliation(s)
- S. Falahi
- Zoonotic Diseases Research Centre, Ilam University of Medical Sciences, Ilam, Iran
| | - A. Abdoli
- Department of Parasitology and Mycology, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - A. Kenarkoohi
- Department of Microbiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|
45
|
Manheim D, Wiȩcek W, Schmit V, Morrison J. Exploring Risks of Human Challenge Trials For COVID-19. RISK ANALYSIS : AN OFFICIAL PUBLICATION OF THE SOCIETY FOR RISK ANALYSIS 2021; 41:710-720. [PMID: 33942351 PMCID: PMC8207107 DOI: 10.1111/risa.13726] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Human challenge trials (HCTs) are a potential method to accelerate development of vaccines and therapeutics. However, HCTs for COVID-19 pose ethical and practical challenges, in part due to the unclear and developing risks. In this article , we introduce an interactive model for exploring some risks of a severe acute respiratory syndrome coronavirus-2 (SARS-COV-2) dosing study, a prerequisite for any COVID-19 challenge trials. The risk estimates we use are based on a Bayesian evidence synthesis model which can incorporate new data on infection fatality risks (IFRs) to patients, and infer rates of hospitalization. The model estimates individual risk, which we then extrapolate to overall mortality and hospitalization risk in a dosing study. We provide a web tool to explore risk under different study designs. Based on the Bayesian model, IFR for someone between 20 and 30 years of age is 15.1 in 100,000, with a 95% uncertainty interval from 11.8 to 19.2, while risk of hospitalization is 130 per 100,000 (100-160). However, risk will be reduced in an HCT via screening for comorbidities, selecting lower-risk population, and providing treatment. Accounting for this with stronger assumptions, we project the fatality risk to be as low as 2.5 per 100,000 (1.6-3.9) and the hospitalization risk to be 22.0 per 100,000 (14.0-33.7). We therefore find a 50-person dosing trial has a 99.74% (99.8-99.9%) chance of no fatalities, and a 98.9% (98.3-99.3%) probability of no cases requiring hospitalization.
Collapse
Affiliation(s)
- David Manheim
- 1DaySooner, Wilmington, DE, USA
- Health and Risk Communication Research Center, University of Haifa, School of Public Health, Haifa, Israel
| | - Witold Wiȩcek
- 1DaySooner, Wilmington, DE, USA
- WAW Statistical Consulting Ltd., UK
| | | | | |
Collapse
|
46
|
Wong SS, Oshansky CM, Guo XZJ, Ralston J, Wood T, Reynolds GE, Seeds R, Jelley L, Waite B, Jeevan T, Zanin M, Widdowson MA, Huang QS, Thomas PG, Webby RJ. Activated CD4 + T cells and CD14 hiCD16 + monocytes correlate with antibody response following influenza virus infection in humans. Cell Rep Med 2021; 2:100237. [PMID: 33948570 PMCID: PMC8080109 DOI: 10.1016/j.xcrm.2021.100237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/25/2021] [Accepted: 03/10/2021] [Indexed: 12/19/2022]
Abstract
The failure to mount an antibody response following viral infection or seroconversion failure is a largely underappreciated and poorly understood phenomenon. Here, we identified immunologic markers associated with robust antibody responses after influenza virus infection in two independent human cohorts, SHIVERS and FLU09, based in Auckland, New Zealand and Memphis, Tennessee, USA, respectively. In the SHIVERS cohort, seroconversion significantly associates with (1) hospitalization, (2) greater numbers of proliferating, activated CD4+ T cells, but not CD8+ T cells, in the periphery during the acute phase of illness, and (3) fewer inflammatory monocytes (CD14hiCD16+) by convalescence. In the FLU09 cohort, fewer CD14hiCD16+ monocytes during early illness in the nasal mucosa were also associated with the generation of influenza-specific mucosal immunoglobulin A (IgA) and IgG antibodies. Our study demonstrates that seroconversion failure after infection is a definable immunological phenomenon, associated with quantifiable cellular markers that can be used to improve diagnostics, vaccine efficacy, and epidemiologic efforts.
Collapse
Affiliation(s)
- Sook-San Wong
- State Key Laboratory for Respiratory Diseases, Guangzhou Medical University, 151 Dongfengxi Road, Yuexiu District, Guangzhou 510000, China
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- School of Public Health, The University of Hong Kong, 7 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Christine M. Oshansky
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), US Department of Health and Human Services (DHHS), 200 C Street, SW, Washington, DC 20201, USA
| | - Xi-Zhi J. Guo
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Jacqui Ralston
- Institute for Environmental Science and Research, NCBID Wallaceville, 66 Ward Street, Upper Hutt 5018, New Zealand
| | - Timothy Wood
- Institute for Environmental Science and Research, NCBID Wallaceville, 66 Ward Street, Upper Hutt 5018, New Zealand
| | - Gary E. Reynolds
- Immunisation Advisory Centre, University of Auckland, Auckland, New Zealand
| | - Ruth Seeds
- Institute for Environmental Science and Research, NCBID Wallaceville, 66 Ward Street, Upper Hutt 5018, New Zealand
- Minsitry for Primary Industries, 66 Ward Street, Upper Hutt 5140, New Zealand
| | - Lauren Jelley
- Institute for Environmental Science and Research, NCBID Wallaceville, 66 Ward Street, Upper Hutt 5018, New Zealand
| | - Ben Waite
- Institute for Environmental Science and Research, NCBID Wallaceville, 66 Ward Street, Upper Hutt 5018, New Zealand
| | - Trushar Jeevan
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Mark Zanin
- State Key Laboratory for Respiratory Diseases, Guangzhou Medical University, 151 Dongfengxi Road, Yuexiu District, Guangzhou 510000, China
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- School of Public Health, The University of Hong Kong, 7 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Marc-Alain Widdowson
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
- Institute of Tropical Medicine (ITM), Nationalestraat 155, 2000 Antwerp, Belgium
| | - Q. Sue Huang
- Institute for Environmental Science and Research, NCBID Wallaceville, 66 Ward Street, Upper Hutt 5018, New Zealand
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Richard J. Webby
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
47
|
Giurgea LT, Park JK, Walters KA, Scherler K, Cervantes-Medina A, Freeman A, Rosas LA, Kash JC, Taubenberger JK, Memoli MJ. The effect of calcium and magnesium on activity, immunogenicity, and efficacy of a recombinant N1/N2 neuraminidase vaccine. NPJ Vaccines 2021; 6:48. [PMID: 33824333 PMCID: PMC8024250 DOI: 10.1038/s41541-021-00310-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/07/2021] [Indexed: 12/26/2022] Open
Abstract
Despite the importance of immunity against neuraminidase (NA), NA content and immunogenicity are neglected in current influenza vaccines. To address this, a recombinant N1/N2 NA vaccine (NAV) was developed. Stability assays were used to determine optimal temperature and buffer conditions for vaccine storage. The effect of divalent cation-related enhancement of NA stability and activity on N1 and N2 immunogenicity and efficacy against viral challenge was assessed. Differences in activity between N1 and N2 and cation-related activity enhancement did not translate into differences in immunogenicity or efficacy. NAV-vaccinated mice showed robust antibody titers against N1 and N2, and after challenge with influenza A (H1N1) virus, decreased viral titers and decreased antiviral and inflammatory responses by transcriptomic analysis. These findings provide guidance for optimal storage and assessment of NA-based vaccines and confirm the importance of NA in influenza vaccination strategies in attenuating viral replication and limiting inflammatory responses necessary to clear infection.
Collapse
Affiliation(s)
- Luca T Giurgea
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Jae-Keun Park
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Adriana Cervantes-Medina
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ashley Freeman
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Luz Angela Rosas
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John C Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffery K Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Matthew J Memoli
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
48
|
Hemmer CJ, Hufert F, Siewert S, Reisinger E. Protection From COVID-19. DEUTSCHES ARZTEBLATT INTERNATIONAL 2021; 118:59-65. [PMID: 33785117 DOI: 10.3238/arztebl.m2021.0119] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 10/26/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Since the beginning of 2020 the SARS-CoV-2 virus has spread to nearly every country in the world. The mainly airborne pathogen has led to large numbers of deaths, principally in elderly and vulnerable segments of the population. Protective vaccines have recently become available, but it is not yet clear whether and when population-wide immunity will be achieved. The existence of evidence for the protective effect of masks covering the mouth and nose is a topic of public debate. METHODS A selective literature search was carried out in PubMed. Data from the German Robert Koch Institute and the Centers for Disease Control and Prevention were also taken into account. RESULTS When talking, as many as 20 000 droplets ranging in size from 20 to 500 μM are released every second. According to PCR tests, the amount of virus exhaled is highest immediately before the onset of symptoms. No randomized trials have been conducted on the effect of masks covering the mouth and nose. A metaanalysis of 29 studies on infection with SARS-CoV-2, SARS, or MERS revealed that type N-95 masks (corresponding approximately to FFP-2), surgical masks, or similar multilayer cotton masks can greatly reduce the infection risk for the wearers (RR 0.34 [0.26; 0.45], with moderate heterogeneity [I2 = 48%]). Model experiments and case reports suggest that masks covering the mouth and nose afford considerable protection against transmission of SARS-CoV-2 and other airborne diseases by reducing release of and exposure to potentially infectious droplets; in addition, infections that do occur take a milder course. A limitation of the studies analyzed is that in most cases, this effect cannot be viewed in isolation from the protective impact of other measures (distancing, hygiene precautions). CONCLUSION It can plausibly be assumed that consistent use of masks covering the mouth and nose can play an important role in containing the spread of SARSCoV- 2.
Collapse
Affiliation(s)
- Christoph Josef Hemmer
- Department of Tropical Medicine and Infectious Diseases, Rostock University Medical CenterDepartment of Microbiology & Virology, Brandenburg Medical School Theodor FontaneInstitute for Implant Technology and Biomaterials (IIB); Rostock University Medical Center
| | | | | | | |
Collapse
|
49
|
George MM, McIntyre CJ, Zhou J, Kugathasan R, Amos DC, Dillon IJ, Barclay WS, Tolley NS. Viral Infectivity in Patients Undergoing Tracheotomy With COVID-19: A Preliminary Study. Otolaryngol Head Neck Surg 2021; 165:819-826. [PMID: 33752547 PMCID: PMC8008096 DOI: 10.1177/01945998211004255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Objective To establish the presence of live virus and its association with polymerase
chain reaction (PCR) positivity and antibody status in patients with
COVID-19 undergoing tracheotomy. Study Design Prospective observational study. Setting Single institution across 3 hospital sites during the first wave of the
COVID-19 pandemic. Methods Patients who were intubated for respiratory wean tracheotomy underwent
SARS-CoV-2 PCR nasal, throat, and endotracheal tube swabs at the time of the
procedure. These were assessed via quantitative real-time reverse
transcription PCR. The tracheal tissue excised during the tracheotomy was
cultured for SARS-CoV-2 with Vero E6 and Caco2 cells. Serum was assessed for
antibody titers against SARS-CoV-2 via neutralization assays. Results Thirty-seven patients were included in this study. The mean number of days
intubated prior to undergoing surgical tracheotomy was 27.8. At the time of
the surgical tracheotomy, PCR swab testing yielded 8 positive results, but
none of the 35 individuals who underwent tissue culture were positive for
SARS-CoV-2. All 18 patients who had serum sampling demonstrated
neutralization antibodies, with a minimum titer of 1:80. Conclusion In our series, irrespective of positive PCR swab, the likelihood of
infectivity during tracheotomy remains low given negative tracheal tissue
cultures. While our results do not undermine national and international
guidance on tracheotomy after day 10 of intubation, given the length of time
to procedure in our data, infectivity at 10 days cannot be excluded. We do
however suggest that a preoperative negative PCR swab not be a prerequisite
and that antibody titer levels may serve as a useful adjunct for assessment
of infectivity.
Collapse
Affiliation(s)
- Manish M George
- Imperial College London, London, UK.,Imperial College NHS Healthcare Trust, London, UK
| | - Charlotte J McIntyre
- Imperial College London, London, UK.,Imperial College NHS Healthcare Trust, London, UK
| | - Jie Zhou
- Imperial College London, London, UK
| | | | - Dora C Amos
- Imperial College NHS Healthcare Trust, London, UK
| | | | | | - Neil S Tolley
- Imperial College London, London, UK.,Imperial College NHS Healthcare Trust, London, UK
| |
Collapse
|
50
|
Noushad M, Al-Saqqaf IS. COVID-19: Is herd immunity the only option for fragile Yemen? Int J Infect Dis 2021; 106:79-82. [PMID: 33737135 PMCID: PMC7959686 DOI: 10.1016/j.ijid.2021.03.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/30/2022] Open
Abstract
The first case of COVID-19 in Yemen was confirmed on 10 April 2020. Having faced with a six-year long conflict that has destroyed half of its healthcare facilities and displaced millions, predictions of infections and mortality in Yemen suggested a looming healthcare catastrophe. Difficulty in implementing coordinated lockdowns and preventive measures due to the daily labor working nature of the majority of the population, provided the perfect breeding ground for the SARS-CoV-2 virus. However, official figures of infections and mortality are very low and there have not been confirmed reports of excess mortality. This could indicate that Yemen is silently marching towards forced herd immunity. Seroprevalence studies will provide useful insight into the COVID-19 transmission trajectory in Yemen, which can serve as a guide in planning vaccine distribution strategies and allocating the limited funds wisely.
Collapse
Affiliation(s)
- Mohammed Noushad
- College of Dentistry, Dar Al Uloom University, Riyadh, Saudi Arabia.
| | | |
Collapse
|