1
|
Sigal A, Neher RA, Lessells RJ. The consequences of SARS-CoV-2 within-host persistence. Nat Rev Microbiol 2025; 23:288-302. [PMID: 39587352 DOI: 10.1038/s41579-024-01125-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/27/2024]
Abstract
SARS-CoV-2 causes an acute respiratory tract infection that resolves in most people in less than a month. Yet some people with severely weakened immune systems fail to clear the virus, leading to persistent infections with high viral titres in the respiratory tract. In a subset of cases, persistent SARS-CoV-2 replication results in an accelerated accumulation of adaptive mutations that confer escape from neutralizing antibodies and enhance cellular infection. This may lead to the evolution of extensively mutated SARS-CoV-2 variants and introduce an element of chance into the timing of variant evolution, as variant formation may depend on evolution in a single person. Whether long COVID is also caused by persistence of replicating SARS-CoV-2 is controversial. One line of evidence is detection of SARS-CoV-2 RNA and proteins in different body compartments long after SARS-CoV-2 infection has cleared from the upper respiratory tract. However, thus far, no replication competent virus has been cultured from individuals with long COVID who are immunocompetent. In this Review, we consider mechanisms of viral persistence, intra-host evolution in persistent infections, the connection of persistent infections with SARS-CoV-2 variants and the possible role of SARS-CoV-2 persistence in long COVID. Understanding persistent infections may therefore resolve much of what is still unclear in COVID-19 pathophysiology, with possible implications for other emerging viruses.
Collapse
Affiliation(s)
- Alex Sigal
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
- Africa Health Research Institute, Durban, South Africa.
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Richard A Neher
- Biozentrum, University of Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Richard J Lessells
- KwaZulu-Natal Research Innovation & Sequencing Platform, School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| |
Collapse
|
2
|
Kozanli E, Winkel AMAM, Han AX, van den Brink S, van den Brandt A, Haverkort ME, Euser SM, Russell CA, de Jong MD, van Houten MA, van Lelyveld SFL, Eggink D. Shortened SARS-CoV-2 Viral RNA Shedding in Saliva During Early Omicron Compared to Wild-Type Pandemic Phase. J Infect Dis 2025; 231:940-945. [PMID: 39823580 DOI: 10.1093/infdis/jiaf031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/20/2024] [Accepted: 01/13/2025] [Indexed: 01/19/2025] Open
Abstract
This study compared the dynamics of SARS-CoV-2 viral shedding in saliva between wild-type virus-infected and Omicron-infected household cohorts. Preexisting immunity in participants likely shortens the viral RNA shedding duration and lowers viral load peaks. Frequent saliva sampling can be a convenient tool to study viral load dynamics.
Collapse
Affiliation(s)
- Eva Kozanli
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Angelique M A M Winkel
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Spaarne Gasthuis Academy, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - Alvin X Han
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sharon van den Brink
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Annemarie van den Brandt
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Mildred E Haverkort
- Department of Infectious Disease Control, Public Health Services Kennemerland, Haarlem, The Netherlands
| | - Sjoerd M Euser
- Spaarne Gasthuis Academy, Spaarne Gasthuis, Hoofddorp, The Netherlands
- Department of Epidemiology and Infection Prevention, Regional Public Health Laboratory Kennemerland, Haarlem, The Netherlands
| | - Colin A Russell
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Menno D de Jong
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Marlies A van Houten
- Spaarne Gasthuis Academy, Spaarne Gasthuis, Hoofddorp, The Netherlands
- Department of Pediatrics, Spaarne Gasthuis, Haarlem and Hoofddorp, The Netherlands
| | - Steven F L van Lelyveld
- Spaarne Gasthuis Academy, Spaarne Gasthuis, Hoofddorp, The Netherlands
- Department of Internal Medicine, Spaarne Gasthuis Hospital, Haarlem/Hoofddorp, The Netherlands
| | - Dirk Eggink
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Bendall EE, Dimcheff D, Papalambros L, Fitzsimmons WJ, Zhu Y, Schmitz J, Halasa N, Chappell J, Martin ET, Biddle JE, Smith-Jeffcoat SE, Rolfes MA, Mellis A, Talbot HK, Grijalva C, Lauring AS. In depth sequencing of a serially sampled household cohort reveals the within-host dynamics of Omicron SARS-CoV-2 and rare selection of novel spike variants. PLoS Pathog 2025; 21:e1013134. [PMID: 40294030 PMCID: PMC12074595 DOI: 10.1371/journal.ppat.1013134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 05/13/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
SARS-CoV-2 has undergone repeated and rapid evolution to circumvent host immunity. However, outside of prolonged infections in immunocompromised hosts, within-host positive selection has rarely been detected. Here we combine daily longitudinal sampling of individuals with replicate sequencing to increase the accuracy of and lower the threshold for variant calling. We sequenced 577 specimens from 105 individuals in a household cohort during the BA.1/BA.2 variant period. Individuals exhibited extremely low viral diversity, and we estimated a low within-host evolutionary rate. Within-host dynamics were dominated by genetic drift and purifying selection. Positive selection was rare but highly concentrated in spike. A Wright Fisher Approximate Bayesian Computational model identified positive selection at 14 loci with 7 in spike, including S:448 and S:339. This detectable immune-mediated selection is unusual in acute respiratory infections and may be caused by the relatively narrow antibody repertoire in individuals during the early Omicron phase of the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Emily E. Bendall
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Derek Dimcheff
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Leigh Papalambros
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - William J. Fitzsimmons
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yuwei Zhu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jonathan Schmitz
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Natasha Halasa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - James Chappell
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Emily T. Martin
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jessica E. Biddle
- Centers for Disease Control and Prevention, Atlanta, GeorgiaUnited States of America
| | | | - Melissa A. Rolfes
- Centers for Disease Control and Prevention, Atlanta, GeorgiaUnited States of America
| | - Alexandra Mellis
- Centers for Disease Control and Prevention, Atlanta, GeorgiaUnited States of America
| | - H. Keipp Talbot
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, Tennessee Tennessee, United States of America
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Carlos Grijalva
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Adam S. Lauring
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
4
|
Deming ME, Brown ER, McArthur MA, Schrag SJ, Arvay M, Humphrys M, Ravel J, Adelglass J, Essink B, Musante DB, Maguire R, Gorman R, Formentini E, Mason R, Robb ML, Neuzil KM, Rapaka RR, Wolff P, Kotloff KL. Vaccine efficacy of NVX-CoV2373 against SARS-CoV-2 infection in adolescents in the USA: an ancillary study to a phase 3, observer-blinded, randomised, placebo-controlled trial. THE LANCET. MICROBE 2025; 6:100984. [PMID: 39884302 DOI: 10.1016/j.lanmic.2024.100984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 02/01/2025]
Abstract
BACKGROUND Although existing COVID-19 vaccines are known to be highly effective against severe disease and death, data are needed to assess their ability to reduce SARS-CoV-2 infection. We aimed to estimate the efficacy of the NVX-CoV2373 protein subunit vaccine against SARS-CoV-2 infection, regardless of symptoms, among adolescents. METHODS We performed an ancillary observational study (SNIFF) to the phase 3, observer-blinded, randomised, placebo-controlled PREVENT-19 trial that assessed vaccine efficacy against symptomatic COVID-19 in the USA. Participants in the PREVENT-19 trial included healthy adolescents aged 12-17 years and with no history of laboratory-confirmed SARS-CoV-2 infection. They were randomly assigned (2:1) to receive either the NVX-CoV2373 (Novavax, Gaithersburg, MD, USA) vaccine (immediate NVX-CoV2373 group) or placebo (delayed NVX-CoV2373 group) on days 0 and 21 (initial series). After 2 months, in a crossover series, participants received two doses, 21 days apart, of the intervention that they did not receive in their initial series. Participants at 47 of the PREVENT-19 sites were invited to participate in the SNIFF study and self-collect nasal swabs at home twice weekly for SARS-CoV-2 testing to assess vaccine efficacy against SARS-CoV-2 infection. This primary outcome was defined as the first identification of SARS-CoV-2 detected by RT-PCR, regardless of symptoms, with onset within 4 weeks after the second dose of the initial vaccination series until the second dose of the crossover series. Secondary outcomes were vaccine efficacy against asymptomatic and minimally symptomatic SARS-CoV-2 infection, durability of vaccine efficacy against SARS-CoV-2 infection, and durability of vaccine efficacy against asymptomatic and minimally symptomatic infections. Outcomes were analysed in the modified intention-to-treat population, which included all participants without previous SARS-CoV-2 infection and was restricted to participants enrolled within 4 weeks of the second dose of the primary (primary analysis population) or crossover (post-crossover analysis population) series. This study is registered with ClinicalTrials.gov (NCT04611802). FINDINGS Between June 1 and Dec 17, 2021, 1196 (53·2%) of the 2247 adolescent participants recruited in the PREVENT-19 trial enrolled in the SNIFF study. The primary analysis population included 471 participants in the immediate NVX-CoV2373 group and 220 in the delayed NVX-CoV2373 group. Incidence of SARS-CoV-2 infection was 14·9 cases per 100 person-years (95% CI 7·9-25·5) in the immediate group and 54·2 cases per 100 person-years (33·6-82·9) in the delayed group; vaccine efficacy was 73·5% (95% CI 47·1-86·7; p=0·0002). Incidence of minimally symptomatic or asymptomatic SARS-CoV-2 infection was 10·3 cases per 100 person-years (95% CI 4·7-19·6) in the immediate group and 36·1 cases per 100 person-years (19·8-60·7) in the delayed group; vaccine efficacy was 72·8% (95% CI 37·1-88·2; p=0·0023). After the second crossover dose, incidence of SARS-CoV-2 was 14·6 cases per 100 person-years (95% CI 8·6-23·0) in the immediate group (receiving placebo at crossover) and 9·1 cases per 100 person-years (3·0-21·3) in the delayed group, with a durability ratio of 160·3 (95% CI 59·5-431·6; p=0·35). Almost all infections after crossover were minimally symptomatic or asymptomatic, with a durability ratio of 151·4 (55·9-410·4; p=0·41). INTERPRETATION Among adolescents participating in the PREVENT-19 trial during the delta (B.1.617.2) variant wave of the COVID-19 pandemic, the NVX-CoV2373 vaccine was highly efficacious against SARS-CoV-2 infection regardless of symptoms, indicating its potential to reduce the reservoir of infections that contribute to community transmission. FUNDING US Department of Health and Human Services, Administration for Strategic Preparedness and Response, Biomedical Advanced Research and Development Authority, National Institute of Allergy and Infectious Diseases, and National Institutes of Health.
Collapse
Affiliation(s)
- Meagan E Deming
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | - Monica A McArthur
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stephanie J Schrag
- US COVID-19 Domestic Response and Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Melissa Arvay
- US COVID-19 Domestic Response and Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Mike Humphrys
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | - Rebecca Maguire
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Richard Gorman
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | - Robin Mason
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Merlin L Robb
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rekha R Rapaka
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Peter Wolff
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Washington, DC, USA
| | - Karen L Kotloff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
5
|
Phan T, Ribeiro RM, Edelstein GE, Boucau J, Uddin R, Marino C, Liew MY, Barry M, Choudhary MC, Tien D, Su K, Reynolds Z, Li Y, Sagar S, Vyas TD, Kawano Y, Sparks JA, Hammond SP, Wallace Z, Vyas JM, Li JZ, Siedner MJ, Barczak AK, Lemieux JE, Perelson AS. Modeling suggests SARS-CoV-2 rebound after nirmatrelvir-ritonavir treatment is driven by target cell preservation coupled with incomplete viral clearance. J Virol 2025; 99:e0162324. [PMID: 39902924 PMCID: PMC11915799 DOI: 10.1128/jvi.01623-24] [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: 09/13/2024] [Accepted: 12/22/2024] [Indexed: 02/06/2025] Open
Abstract
In a subset of SARS-CoV-2-infected individuals treated with the antiviral nirmatrelvir-ritonavir, the virus rebounds following treatment. The mechanisms driving this rebound are not well understood. We used a mathematical model to describe the longitudinal viral load dynamics of 51 individuals treated with nirmatrelvir-ritonavir, 20 of whom rebounded. Target cell preservation, either by a robust innate immune response or initiation of N-R near the time of symptom onset, coupled with incomplete viral clearance, appears to be the main factor leading to viral rebound. Moreover, the occurrence of viral rebound is likely influenced by the time of treatment initiation relative to the progression of the infection, with earlier treatments leading to a higher chance of rebound. A comparison with an untreated cohort suggests that early treatments with nirmatrelvir-ritonavir may be associated with a delay in the onset of an adaptive immune response. Nevertheless, our model demonstrates that extending the course of nirmatrelvir-ritonavir treatment to a 10-day regimen may greatly diminish the chance of rebound in people with mild-to-moderate COVID-19 and who are at high risk of progression to severe disease. Altogether, our results suggest that in some individuals, a standard 5-day course of nirmatrelvir-ritonavir starting around the time of symptom onset may not completely eliminate the virus. Thus, after treatment ends, the virus can rebound if an effective adaptive immune response has not fully developed. These findings on the role of target cell preservation and incomplete viral clearance also offer a possible explanation for viral rebounds following other antiviral treatments for SARS-CoV-2. IMPORTANCE Nirmatrelvir-ritonavir is an effective treatment for SARS-CoV-2. In a subset of individuals treated with nirmatrelvir-ritonavir, the initial reduction in viral load is followed by viral rebound once treatment is stopped. We show that the timing of treatment initiation with nirmatrelvir-ritonavir may influence the risk of viral rebound. Nirmatrelvir-ritonavir stops viral growth and preserves target cells but may not lead to full clearance of the virus. Thus, once treatment ends, if an effective adaptive immune response has not adequately developed, the remaining virus can lead to rebound. Our results provide insights into the mechanisms of rebound and can help develop better treatment strategies to minimize this possibility.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Ruy M. Ribeiro
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Gregory E. Edelstein
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julie Boucau
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Rockib Uddin
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Caitlin Marino
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - May Y. Liew
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mamadou Barry
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Manish C. Choudhary
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dessie Tien
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Karry Su
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zahra Reynolds
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yijia Li
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Shruti Sagar
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tammy D. Vyas
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yumeko Kawano
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey A. Sparks
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah P. Hammond
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zachary Wallace
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jatin M. Vyas
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan Z. Li
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark J. Siedner
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Africa Health Research Institute, KwaZulu-Natal, South Africa
| | - Amy K. Barczak
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob E. Lemieux
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Alan S. Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
- Santa Fe Institute, Santa Fe, New Mexico, USA
| |
Collapse
|
6
|
Schuh AJ, Amman BR, Guito JC, Graziano JC, Sealy TK, Towner JS. Modeling natural coinfection in a bat reservoir shows modulation of Marburg virus shedding and spillover potential. PLoS Pathog 2025; 21:e1012901. [PMID: 40096181 PMCID: PMC11978059 DOI: 10.1371/journal.ppat.1012901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 04/08/2025] [Accepted: 01/12/2025] [Indexed: 03/19/2025] Open
Abstract
The Egyptian rousette bat (ERB) is a natural reservoir for Marburg virus (MARV; family Filoviridae), a putative reservoir for Sosuga virus (SOSV; family Paramyxoviridae), and a vertebrate reservoir for Kasokero virus (KASV; family Orthonairoviridae); however, the effect of naturally occurring coinfection by those viruses on MARV shedding and spillover potential is unknown. To answer this question, we experimentally infected one cohort of captive-bred ERBs with SOSV+MARV (n=12 bats) or MARV only (n=12 bats) and a second cohort with KASV+MARV (n=12 bats) or MARV only (n=12 bats), and then collected blood, oral swab, and rectal swab specimens throughout the course of infection to monitor viral shedding. Compared to the MARV-monoinfected bat group, the SOSV+MARV-coinfected bat group exhibited a significantly shortened duration of MARV oral shedding and a significantly decreased anti-MARV IgG response, which may increase the capacity for MARV reinfection. In contrast, relative to the MARV-monoinfected bat group, the KASV+MARV-coinfected bat group exhibited significantly increased peak magnitudes and durations of MARV viremia and oral shedding, as well as a significantly increased anti-MARV IgG response. Correspondingly, cumulative MARV shedding loads, a measure of infectiousness, were significantly higher in the KASV+MARV-coinfected bat group than the MARV-monoinfected bat group. Four of the KASV+MARV-coinfected bats were classified as MARV supershedders, together accounting for 72.5% of the KASV-MARV experimental cohort's total shedding. Our results demonstrate that SOSV+MARV and KASV+MARV coinfection of ERBs differentially modulates MARV shedding and anti-MARV IgG responses, thereby implicating MARV coinfection as playing a critical role in bat-to-bat MARV transmission dynamics and spillover potential.
Collapse
Affiliation(s)
- Amy J. Schuh
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- United States Public Health Service Commissioned Corps, Rockville, Maryland, United States of America
| | - Brian R. Amman
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jonathan C. Guito
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - James C. Graziano
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Tara K. Sealy
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jonathan S. Towner
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
7
|
Woodbridge Y, Mandel M, Goldberg Y, Huppert A. Estimating Mean Viral Load Trajectory From Intermittent Longitudinal Data and Unknown Time Origins. Stat Med 2025; 44:e70033. [PMID: 39995297 PMCID: PMC11851093 DOI: 10.1002/sim.70033] [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: 04/19/2024] [Revised: 01/26/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025]
Abstract
Viral load (VL) in the respiratory tract is the leading proxy for assessing infectiousness potential. Understanding the dynamics of disease-related VL within the host is of great importance, as it helps to determine different policies and health recommendations. However, normally the VL is measured on individuals only once, in order to confirm infection, and furthermore, the infection date is unknown. It is therefore necessary to develop statistical approaches to estimate the typical VL trajectory. We show here that, under plausible parametric assumptions, two measures of VL on infected individuals can be used to accurately estimate the VL mean function. Specifically, we consider a discrete-time likelihood-based approach to modeling and estimating partial observed longitudinal samples. We study a multivariate normal model for a function of the VL that accounts for possible correlation between measurements within individuals. We derive an expectation-maximization (EM) algorithm which treats the unknown time origins and the missing measurements as latent variables. Our main motivation is the reconstruction of the daily mean VL, given measurements on patients whose VLs were measured multiple times on different days. Such data should and can be obtained at the beginning of a pandemic with the specific goal of estimating the VL dynamics. For demonstration purposes, the method is applied to SARS-Cov-2 cycle-threshold-value data collected in Israel.
Collapse
Affiliation(s)
- Yonatan Woodbridge
- The Gertner Institute for Epidemiology & Health Policy Research, Sheba Medical CenterRamat GanIsrael
- Department of Computer ScienceHolon Institute of TechnologyHolonIsrael
| | - Micha Mandel
- Department of Statistics and Data ScienceThe Hebrew University of JerusalemJerusalemIsrael
| | - Yair Goldberg
- Faculty of Industrial Engineering and ManagementTechnion ‐ Israel Institute of TechnologyHaifaIsrael
| | - Amit Huppert
- The Gertner Institute for Epidemiology & Health Policy Research, Sheba Medical CenterRamat GanIsrael
- Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medical and Health SciencesTel‐Aviv UniversityTel‐AvivIsrael
| |
Collapse
|
8
|
Porter MK, Viloria Winnett A, Hao L, Shelby N, Reyes JA, Schlenker NW, Romano AE, Tognazzini C, Feaster M, Goh YY, Gale Jr M, Ismagilov RF. The ratio between SARS-CoV-2 RNA viral load and culturable viral titre differs depending on the stage of infection: a case study of household transmission in an adult male. Access Microbiol 2025; 7:000732.v3. [PMID: 39967741 PMCID: PMC11833051 DOI: 10.1099/acmi.0.000732.v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Effective public health measures for communicable diseases rely on the ability to identify infectious individuals and prevent transmission from those individuals. For severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the presence of replication-competent virus in specimens from an individual is the gold standard for confirming infectiousness. However, viral culture from clinical specimens is difficult and infrequently performed. Instead, infectiousness may be inferred based on the abundance of viral RNA (or viral load) in a specimen, which is more easily assessed. For this reason, understanding the relationship between RNA viral load and infectious viral titre has important implications for public health strategy. In this case report, we quantified incident, longitudinal SARS-CoV-2 viral loads collected from saliva and nasal-swab specimens, and viral titre from nasal-swab specimens. We observed that the relationship between viral load and viral titre decreases by over five orders of magnitude throughout the course of the infection. Our work demonstrates the potential for infectious virus even in specimens with low viral loads collected during the early phases of infection.
Collapse
Affiliation(s)
- Michael K. Porter
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Alexander Viloria Winnett
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Linhui Hao
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Natasha Shelby
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jessica A. Reyes
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Noah W. Schlenker
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Anne E. Romano
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | | - Ying-Ying Goh
- Pasadena Public Health Department, Pasadena, CA 91125, USA
| | - Michael Gale Jr
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Rustem F. Ismagilov
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
9
|
Jeong YD, Ejima K, Kim KS, Iwanami S, Hart WS, Thompson RN, Jung IH, Iwami S, Ajelli M, Aihara K. A modeling study to define guidelines for antigen screening in schools and workplaces to mitigate COVID-19 outbreaks. COMMUNICATIONS MEDICINE 2025; 5:2. [PMID: 39753869 PMCID: PMC11699287 DOI: 10.1038/s43856-024-00716-3] [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: 07/13/2023] [Accepted: 12/17/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND In-person interaction offers invaluable benefits to people. To guarantee safe in-person activities during a COVID-19 outbreak, effective identification of infectious individuals is essential. In this study, we aim to analyze the impact of screening with antigen tests in schools and workplaces on identifying COVID-19 infections. METHODS We assess the effectiveness of various screening test strategies with antigen tests in schools and workplaces through quantitative simulations. The primary outcome of our analyses is the proportion of infected individuals identified. The transmission process at the population level is modeled using a deterministic compartmental model. Infected individuals are identified through screening tests or symptom development. The time-varying sensitivity of antigen tests and infectiousness is determined by a viral dynamics model. Screening test strategies are characterized by the screening schedule, sensitivity of antigen tests, screening duration, timing of screening initiation, and available tests per person. RESULTS Here, we show that early and frequent screening is the key to maximizing the effectiveness of the screening program. For example, 44.5% (95% CI: 40.8-47.5) of infected individuals are identified by daily testing, whereas it is only 33.7% (95% CI: 30.5-37.3) when testing is performed at the end of the program duration. If high sensitivity antigen tests (Detection limit: 6.3 × 10 4 copies/mL) are deployed, it reaches 69.3% (95% CI: 66.5-72.5). CONCLUSIONS High sensitivity antigen tests, high frequency screening tests, and immediate initiation of screening tests are important to safely restart educational and economic activities in-person. Our computational framework is useful for assessing screening programs by incorporating situation-specific factors.
Collapse
Affiliation(s)
- Yong Dam Jeong
- Interdisciplinary Biology Laboratory (iBLab), Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
- Department of Mathematics, Pusan National University, Busan, South Korea
| | - Keisuke Ejima
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| | - Kwang Su Kim
- Interdisciplinary Biology Laboratory (iBLab), Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
- Department of Scientific Computing, Pukyong National University, Busan, South Korea
| | - Shoya Iwanami
- Interdisciplinary Biology Laboratory (iBLab), Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - William S Hart
- Mathematical Institute, University of Oxford, Oxford, UK
| | | | - Il Hyo Jung
- Department of Mathematics, Pusan National University, Busan, South Korea
- Finace Fishery Manufacture Industrial Mathematics Center on Big Data, Pusan National University, Busan, South Korea
| | - Shingo Iwami
- Interdisciplinary Biology Laboratory (iBLab), Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan.
- Institute of Mathematics for Industry, Kyushu University, Fukuoka, Japan.
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.
- NEXT-Ganken Program, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan.
- Science Groove Inc., Fukuoka, Japan.
| | - Marco Ajelli
- Laboratory for Computational Epidemiology and Public Health Department of Epidemiology and Biostatistics, Indiana University School of Public Health-, Bloomington, IN, USA
| | - Kazuyuki Aihara
- International Research Center for Neurointelligence, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
10
|
Chua HK, Singh A, Wang Y, Goh YS, Chan CEZ, Chavatte J, Lin RVTP, Su YCF, Ajelli M, Chia PY, Ong SWX, Lye DC, Young BE, Ejima K. Defining the Critical Requisites for Accurate Simulation of SARS-CoV-2 Viral Dynamics: Patient Characteristics and Data Collection Protocol. J Med Virol 2025; 97:e70174. [PMID: 39817600 PMCID: PMC11736999 DOI: 10.1002/jmv.70174] [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: 06/20/2024] [Revised: 11/05/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025]
Abstract
Mathematical models of viral dynamics are crucial in understanding infection trajectories. However, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral load data often includes limited sparse observations with significant heterogeneity. This study aims to: (1) understand the impact of patient characteristics in shaping the temporal viral load trajectory and (2) establish a data collection protocol (DCP) to reliably reconstruct individual viral load trajectories. We collected longitudinal viral load data for SARS-CoV-2 Delta and Omicron variants from 243 patients in Singapore (2021-2022). A viral dynamics model was calibrated using patients' age, symptom presence, and vaccination status. We accessed associations between these patient characteristics and aspects of viral dynamics using linear regression models. We evaluated the accuracy of viral load trajectory estimation under different simulated DCPs by varying patient numbers, test frequencies, and test intervals. Older unvaccinated individuals had a longer viral shedding duration due to lower infection and cell death rates. Higher peak viral loads were found in older, symptomatic, and vaccinated individuals, with earlier peaks in younger vaccinated individuals. Symptom presence and vaccination resulted in a shorter time from infection to diagnosis. To accurately estimate viral dynamics, more frequent tests, longer test intervals, and larger patient samples are required. For 500 patients, a 21-day follow-up with measurements every 3 days and an 8-day follow-up with daily measurements was optimal for the Delta and Omicron variants, respectively. Patient characteristics significantly impacted viral dynamics. Our analytic approach and recommended DCPs can enhance preparedness and response to emerging pathogens beyond SARS-CoV-2.
Collapse
Affiliation(s)
- Hoong Kai Chua
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - Ananya Singh
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Yuqian Wang
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Yun Shan Goh
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | | | | | | | - Yvonne C. F. Su
- Programme in Emerging Infectious Diseases, Duke‐NUS Medical SchoolSingaporeSingapore
| | - Marco Ajelli
- Laboratory for Computational Epidemiology and Public Health, Department of Epidemiology and BiostatisticsIndiana University School of Public Health‐BloomingtonBloomingtonIndianaUSA
| | - Po Ying Chia
- National Centre for Infectious DiseasesSingaporeSingapore
- Department of Infectious DiseasesTan Tock Seng HospitalSingaporeSingapore
| | - Sean W. X. Ong
- National Centre for Infectious DiseasesSingaporeSingapore
- Department of Infectious DiseasesTan Tock Seng HospitalSingaporeSingapore
| | - David Chien Lye
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
- National Centre for Infectious DiseasesSingaporeSingapore
- Department of Infectious DiseasesTan Tock Seng HospitalSingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Barnaby E. Young
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
- National Centre for Infectious DiseasesSingaporeSingapore
- Department of Infectious DiseasesTan Tock Seng HospitalSingaporeSingapore
| | - Keisuke Ejima
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
- National Centre for Infectious DiseasesSingaporeSingapore
| |
Collapse
|
11
|
Fu J, Mo R, Li Z, Xu S, Cheng X, Lu B, Shi S. An extraction-free one-pot assay for rapid detection of Klebsiella pneumoniae by combining RPA and CRISPR/Cas12a. Biosens Bioelectron 2025; 267:116740. [PMID: 39244837 DOI: 10.1016/j.bios.2024.116740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Klebsiella pneumoniae poses a significant threat to global public health. Traditional clinical diagnostic methods, such as bacterial culture and microscopic identification, are not suitable for point-of-care testing. In response, based on the suboptimal protospacer adjacent motifs, this study develops an extraction-free one-pot assay, named EXORCA (EXtraction-free One-pot RPA-CRISPR/Cas12a assay), designed for the immediate, sensitive and efficient detection of K. pneumoniae. The EXORCA assay can be completed within approximately 30 min at a constant temperature and allows for the visualization of results either through a fluorescence reader or directly by the naked eye under blue light. The feasibility of the assay was evaluated using twenty unextracted clinical samples, achieving a 100% (5/5) positive predictive value and a 100% (15/15) negative predictive value in comparison to qPCR. These results suggest that the EXORCA assay holds significant potential as a point-of-care testing tool for the rapid identification of pathogens, such as K. pneumonia.
Collapse
Affiliation(s)
- Jinyu Fu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Rurong Mo
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Ziyao Li
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China; Changping Laboratory, Beijing, China; Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Shijie Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Xiyu Cheng
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing, 100044, China
| | - Binghuai Lu
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China; Changping Laboratory, Beijing, China; Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuobo Shi
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| |
Collapse
|
12
|
Naderi S, Sagan SM, Shapiro BJ. Within-host genetic diversity of SARS-CoV-2 across animal species. Virus Evol 2024; 11:veae117. [PMID: 39830312 PMCID: PMC11739616 DOI: 10.1093/ve/veae117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/19/2024] [Accepted: 12/30/2024] [Indexed: 01/22/2025] Open
Abstract
Infectious disease transmission to different host species makes eradication very challenging and expands the diversity of evolutionary trajectories taken by the pathogen. Since the beginning of the ongoing COVID-19 pandemic, SARS-CoV-2 has been transmitted from humans to many different animal species, in which viral variants of concern could potentially evolve. Previously, using available whole genome consensus sequences of SARS-CoV-2 from four commonly sampled animals (mink, deer, cat, and dog), we inferred similar numbers of transmission events from humans to each animal species. Using a genome-wide association study, we identified 26 single nucleotide variants (SNVs) that tend to occur in deer-more than any other animal-suggesting a high rate of viral adaptation to deer. The reasons for this rapid adaptive evolution remain unclear, but within-host evolution-the ultimate source of the viral diversity that transmits globally-could provide clues. Here, we quantify intra-host SARS-CoV-2 genetic diversity across animal species and show that deer harbor more intra-host SNVs (iSNVs) than other animals, providing a larger pool of genetic diversity for natural selection to act upon. Mixed infections involving more than one viral lineage are unlikely to explain the higher diversity within deer. Rather, a combination of higher mutation rates, longer infections, and species-specific selective pressures are likely explanations. Combined with extensive deer-to-deer transmission, the high levels of within-deer viral diversity help explain the apparent rapid adaptation of SARS-CoV-2 to deer.
Collapse
Affiliation(s)
- Sana Naderi
- Department of Microbiology and Immunology, McGill University, 3775 University Street Montreal, QC H3A 2B4, Canada
| | - Selena M Sagan
- Department of Microbiology and Immunology, McGill University, 3775 University Street Montreal, QC H3A 2B4, Canada
- Department of Microbiology and Immunology, The University of British Columbia, 2350 Health Sciences Mall #1365 Vancouver, BC V6T 1Z3, Canada
| | - B. Jesse Shapiro
- Department of Microbiology and Immunology, McGill University, 3775 University Street Montreal, QC H3A 2B4, Canada
- McGill Genome Centre, 740 avenue Dr Penfield Montreal, QC H3A 0G1, Canada
| |
Collapse
|
13
|
Mohammadi A, Chiang S, Li F, Wei F, Lau CS, Aziz M, Ibarrondo FJ, Fulcher JA, Yang OO, Chia D, Kim Y, Wong DTW. Direct detection of 4-dimensions of SARS-CoV-2: infection (vRNA), infectivity (antigen), binding antibody, and functional neutralizing antibody in saliva. Sci Rep 2024; 14:30792. [PMID: 39730575 DOI: 10.1038/s41598-024-81019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 11/22/2024] [Indexed: 12/29/2024] Open
Abstract
We developed a 4-parameter clinical assay using Electric Field Induced Release and Measurement (EFIRM) technology to simultaneously assess SARS-CoV-2 RNA (vRNA), nucleocapsid antigen, host binding (BAb) and neutralizing antibody (NAb) levels from a drop of saliva with performance that equals or surpasses current EUA-approved tests. The vRNA and antigen assays achieved lower limit of detection (LOD) of 100 copies/reaction and 3.5 TCID₅₀/mL, respectively. The vRNA assay differentiated between acutely infected (n = 10) and infection-naïve patients (n = 33) with an AUC of 0.9818, sensitivity of 90%, and specificity of 100%. The antigen assay similarly differentiated these patient populations with an AUC of 1.000. The BAb assay detected BAbs with an LOD of 39 pg/mL and distinguished acutely infected (n = 35), vaccinated with prior infection (n = 13), and vaccinated infection-naïve patients (n = 13) from pre-pandemic (n = 81) with AUC of 0.9481, 1.000, and 0.9962, respectively. The NAb assay detected NAbs with a LOD of 31.6 Unit/mL and differentiated between COVID-19 recovered or vaccinated patients (n = 31) and pre-pandemic controls (n = 60) with an AUC 0.923, sensitivity of 87.10%, and specificity of 86.67%. Our combo assay represents a significant technological advancement to simultaneously address SARS-CoV-2 infection and immunity, and it lays the foundation for tackling potential future pandemics.
Collapse
Grants
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
Collapse
Affiliation(s)
- Aida Mohammadi
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Samantha Chiang
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Feng Li
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Fang Wei
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | | | - Mohammad Aziz
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Francisco J Ibarrondo
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer A Fulcher
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Otto O Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - David Chia
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Yong Kim
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA.
| | - David T W Wong
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA.
| |
Collapse
|
14
|
Bendall EE, Dimcheff D, Papalambros L, Fitzsimmons WJ, Zhu Y, Schmitz J, Halasa N, Chappell J, Martin ET, Biddle JE, Smith-Jeffcoat SE, Rolfes MA, Mellis A, Talbot HK, Grijalva C, Lauring AS. In depth sequencing of a serially sampled household cohort reveals the within-host dynamics of Omicron SARS-CoV-2 and rare selection of novel spike variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.21.624722. [PMID: 39605326 PMCID: PMC11601520 DOI: 10.1101/2024.11.21.624722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
SARS-CoV-2 has undergone repeated and rapid evolution to circumvent host immunity. However, outside of prolonged infections in immunocompromised hosts, within-host positive selection has rarely been detected. The low diversity within-hosts and strong genetic linkage among genomic sites make accurately detecting positive selection difficult. Longitudinal sampling is a powerful method for detecting selection that has seldom been used for SARS-CoV-2. Here we combine longitudinal sampling with replicate sequencing to increase the accuracy of and lower the threshold for variant calling. We sequenced 577 specimens from 105 individuals from a household cohort primarily during the BA.1/BA.2 variant period. There was extremely low diversity and a low rate of divergence. Specimens had 0-12 intrahost single nucleotide variants (iSNV) at >0.5% frequency, and the majority of the iSNV were at frequencies <2%. Within-host dynamics were dominated by genetic drift and purifying selection. Positive selection was rare but highly concentrated in spike. Two individuals with BA.1 infections had S:371F, a lineage defining substitution for BA.2. A Wright Fisher Approximate Bayesian Computational model identified positive selection at 14 loci with 7 in spike, including S:448 and S:339. We also detected significant genetic hitchhiking between synonymous changes and nonsynonymous iSNV under selection. The detectable immune-mediated selection may be caused by the relatively narrow antibody repertoire in individuals during the early Omicron phase of the SARS-CoV-2 pandemic. As both the virus and population immunity evolve, understanding the corresponding shifts in SARS-CoV-2 within-host dynamics will be important.
Collapse
Affiliation(s)
- Emily E. Bendall
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Derek Dimcheff
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Leigh Papalambros
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Yuwei Zhu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan Schmitz
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Natasha Halasa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James Chappell
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Emily T. Martin
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | - H. Keipp Talbot
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carlos Grijalva
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adam S. Lauring
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
15
|
Iyaniwura SA, Ribeiro RM, Zitzmann C, Phan T, Ke R, Perelson AS. The kinetics of SARS-CoV-2 infection based on a human challenge study. Proc Natl Acad Sci U S A 2024; 121:e2406303121. [PMID: 39508770 PMCID: PMC11573497 DOI: 10.1073/pnas.2406303121] [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: 03/28/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
Studying the early events that occur after viral infection in humans is difficult unless one intentionally infects volunteers in a human challenge study. Here, we use data about severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in such a study in combination with mathematical modeling to gain insights into the relationship between the amount of virus in the upper respiratory tract and the immune response it generates. We propose a set of dynamic models of increasing complexity to dissect the roles of target cell limitation, innate immunity, and adaptive immunity in determining the observed viral kinetics. We introduce an approach for modeling the effect of humoral immunity that describes a decline in infectious virus after immune activation. We fit our models to viral load and infectious titer data from all the untreated infected participants in the study simultaneously. We found that a power-law with a power h < 1 describes the relationship between infectious virus and viral load. Viral replication at the early stage of infection is rapid, with a doubling time of ~2 h for viral RNA and ~3 h for infectious virus. We estimate that adaptive immunity is initiated ~7 to 10 d postinfection and appears to contribute to a multiphasic viral decline experienced by some participants; the viral rebound experienced by other participants is consistent with a decline in the interferon response. Altogether, we quantified the kinetics of SARS-CoV-2 infection, shedding light on the early dynamics of the virus and the potential role of innate and adaptive immunity in promoting viral decline during infection.
Collapse
Affiliation(s)
- Sarafa A Iyaniwura
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Ruy M Ribeiro
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Carolin Zitzmann
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Tin Phan
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Ruian Ke
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Alan S Perelson
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| |
Collapse
|
16
|
Byrne C, Schiffer JT. Ensemble modeling of SARS-CoV-2 immune dynamics in immunologically naïve rhesus macaques predicts that potent, early innate immune responses drive viral elimination. Front Immunol 2024; 15:1426016. [PMID: 39575237 PMCID: PMC11578959 DOI: 10.3389/fimmu.2024.1426016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Introduction An unprecedented breadth of longitudinal viral and multi-scale immunological data has been gathered during SARS-CoV-2 infection. However, due to the high complexity, non-linearity, multi-dimensionality, mixed anatomic sampling, and possible autocorrelation of available immune data, it is challenging to identify the components of the innate and adaptive immune response that drive viral elimination. Novel mathematical models and analytical approaches are required to synthesize contemporaneously gathered cytokine, transcriptomic, flow cytometry, antibody response, and viral load data into a coherent story of viral control, and ultimately to discriminate drivers of mild versus severe infection. Methods We investigated a dataset describing innate, SARS-CoV-2 specific T cell, and antibody responses in the lung during early and late stages of infection in immunologically naïve rhesus macaques. We used multi-model inference and ensemble modeling approaches from ecology and weather forecasting to compare and combine various competing models. Results and discussion Model outputs suggest that the innate immune response plays a crucial role in controlling early infection, while SARS-CoV-2 specific CD4+ T cells correspond to later viral elimination, and anti-spike IgG antibodies do not impact viral dynamics. Among the numerous genes potentially contributing to the innate response, we identified IFI27 as most closely linked to viral load decline. A 90% knockdown of the innate response from our validated model resulted in a ~10-fold increase in peak viral load during infection. Our approach provides a novel methodological framework for future analyses of similar complex, non-linear multi-component immunologic data sets.
Collapse
Affiliation(s)
| | - Joshua T. Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center,
Seattle, WA, United States
| |
Collapse
|
17
|
Schuh L, Markov PV, Voulgaridi I, Bogogiannidou Z, Mouchtouri VA, Hadjichristodoulou C, Stilianakis NI. Within-host dynamics of antiviral treatment with remdesivir for SARS-CoV-2 infection. J R Soc Interface 2024; 21:20240536. [PMID: 39592013 PMCID: PMC11597410 DOI: 10.1098/rsif.2024.0536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/09/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
The effectiveness of antiviral treatment with remdesivir against COVID-19 has been investigated in clinical trials suggesting earlier recovery. However, this effect seems to be rather modest. In this study, we tracked the clinical course of SARS-CoV-2 infections in 369 COVID-19 individuals across a spectrum of illness severities, including both untreated individuals and individuals who received antiviral treatment with remdesivir. Moreover, using a process-based mathematical model, we quantified and analysed the within-host infection dynamics of a total of 88 individuals, of which 69 were untreated and 19 antiviral-treated individuals. For untreated individuals, we found that those hospitalized exhibit lower levels of early immune response and higher cumulative viral loads than those who were not. For treated individuals, we found that those who died were on average hospitalized later after symptom onset than those who survived, underscoring the importance of early medical intervention for severe COVID-19. Finally, our model estimates a rather limited antiviral activity of remdesivir. Our results provide valuable insights into the clinical course of COVID-19 during antiviral treatment with remdesivir and suggest the need for alternative treatment regimens.
Collapse
Affiliation(s)
- Lea Schuh
- Joint Research Centre (JRC), European Commission, Ispra, Italy
| | - Peter V. Markov
- Joint Research Centre (JRC), European Commission, Ispra, Italy
- London School of Hygiene & Tropical Medicine, University of London, London, UK
| | - Ioanna Voulgaridi
- Laboratory of Hygiene and Epidemiology, University of Thessaly, Larissa, Greece
| | | | | | | | - Nikolaos I. Stilianakis
- Joint Research Centre (JRC), European Commission, Ispra, Italy
- Department of Biometry and Epidemiology, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
18
|
Lopez Quezada L, Mba Medie F, Luu RJ, Gaibler RB, Gabriel EP, Rubio LD, Mulhern TJ, Marr EE, Borenstein JT, Fisher CR, Gard AL. Predicting Clinical Outcomes of SARS-CoV-2 Drug Efficacy with a High-Throughput Human Airway Microphysiological System. Adv Biol (Weinh) 2024; 8:e2300511. [PMID: 39123296 DOI: 10.1002/adbi.202300511] [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] [Revised: 12/23/2023] [Indexed: 08/12/2024]
Abstract
The average cost to bring a new drug from its initial discovery to a patient's bedside is estimated to surpass $2 billion and requires over a decade of research and development. There is a need for new drug screening technologies that can parse drug candidates with increased likelihood of clinical utility early in development in order to increase the cost-effectiveness of this pipeline. For example, during the COVID-19 pandemic, resources were rapidly mobilized to identify effective therapeutic treatments but many lead antiviral compounds failed to demonstrate efficacy when progressed to human trials. To address the lack of predictive preclinical drug screening tools, PREDICT96-ALI, a high-throughput (n = 96) microphysiological system (MPS) that recapitulates primary human tracheobronchial tissue,is adapted for the evaluation of differential antiviral efficacy of native SARS-CoV-2 variants of concern. Here, PREDICT96-ALI resolves both the differential viral kinetics between variants and the efficacy of antiviral compounds over a range of drug doses. PREDICT96-ALI is able to distinguish clinically efficacious antiviral therapies like remdesivir and nirmatrelvir from promising lead compounds that do not show clinical efficacy. Importantly, results from this proof-of-concept study track with known clinical outcomes, demonstrate the feasibility of this technology as a prognostic drug discovery tool.
Collapse
Affiliation(s)
| | | | - Rebeccah J Luu
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| | | | | | - Logan D Rubio
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| | | | | | | | | | - Ashley L Gard
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| |
Collapse
|
19
|
Toth DJA, Sheets TR, Beams AB, Ahmed SM, Seegert N, Love J, Keegan LT, Samore MH. Model-based estimates of age-structured SARS-CoV-2 epidemiology in households. BMC Public Health 2024; 24:2965. [PMID: 39455984 PMCID: PMC11515260 DOI: 10.1186/s12889-024-20308-z] [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: 05/02/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Understanding how infectious disease transmission varies from person to person, including associations with age and contact behavior, can help design effective control strategies. Within households, transmission may be highly variable because of differing transmission risks by age, household size, and individual contagiousness. Our aim was to disentangle those factors by fitting mathematical models to SARS-CoV-2 household survey and serologic data. METHODS We surveyed members of 3,381 Utah households from January-April 2021 and performed SARS-CoV-2 antibody testing on all available members. We paired these data with a probabilistic model of household importation and transmission composed of a novel combination of transmission variability and age- and size-structured heterogeneity. We calculated maximum likelihood estimates of mean and variability of household transmission probability between household members in different age groups and different household sizes, simultaneously with importation probability and probabilities of false negative and false positive test results. RESULTS 12.8% of individual participants, residing in 17.4% of the participating households, showed serologic evidence of prior infection or reported a prior positive test on the survey. Serologically positive individuals in younger age groups were less likely than older adults to have tested positive during their infection according to our survey results. Our model results suggested that adolescents and young adults (ages 13-24) acquired SARS-CoV-2 infection outside the household at a rate substantially higher than younger children and older adults. Our estimate of the household secondary attack rate (HSAR) among adults aged 45 and older exceeded HSARs to and/or from younger age groups at a given household size. We found lower HSAR in households with more members, independent of age differences. The age-specific HSAR patterns we found could not be explained by age-dependent biological susceptibility and transmissibility alone, suggesting that age groups contacted each other at different rates within households. CONCLUSIONS We disentangled several factors contributing to age-specific infection risk, including non-household exposure, within-household exposure to specific age groups, and household size. Within-household contact rate differences played a significant role in driving household transmission epidemiology. These findings provide nuanced insights for understanding community outbreak patterns and mechanisms of differential infection risk.
Collapse
Affiliation(s)
- Damon J A Toth
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America.
- Department of Veterans Affairs Salt Lake City Healthcare System, Salt Lake City, UT, United States of America.
- Department of Mathematics, University of Utah, Salt Lake City, UT, United States of America.
| | - Theresa R Sheets
- Department of Mathematics, University of Utah, Salt Lake City, UT, United States of America
| | - Alexander B Beams
- Department of Mathematics, University of Utah, Salt Lake City, UT, United States of America
- Present address: Department of Mathematics, Simon Fraser University, Burnaby, BC, Canada
| | - Sharia M Ahmed
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Nathan Seegert
- Department of Finance, University of Utah David Eccles School of Business, Salt Lake City, UT, United States of America
| | - Jay Love
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Lindsay T Keegan
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Department of Veterans Affairs Salt Lake City Healthcare System, Salt Lake City, UT, United States of America
| | - Matthew H Samore
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Department of Veterans Affairs Salt Lake City Healthcare System, Salt Lake City, UT, United States of America
| |
Collapse
|
20
|
McKee CD, Yu EX, Garcia A, Jackson J, Koyuncu A, Rose S, Azman AS, Lobner K, Sacks E, Van Kerkhove MD, Gurley ES. Superspreading of SARS-CoV-2: a systematic review and meta-analysis of event attack rates and individual transmission patterns. Epidemiol Infect 2024; 152:e121. [PMID: 39377138 PMCID: PMC11488467 DOI: 10.1017/s0950268824000955] [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: 01/23/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 10/09/2024] Open
Abstract
SARS-CoV-2 superspreading occurs when transmission is highly efficient and/or an individual infects many others, contributing to rapid spread. To better quantify heterogeneity in SARS-CoV-2 transmission, particularly superspreading, we performed a systematic review of transmission events with data on secondary attack rates or contact tracing of individual index cases published before September 2021 prior to the emergence of variants of concern and widespread vaccination. We reviewed 592 distinct events and 9,883 index cases from 491 papers. A meta-analysis of secondary attack rates identified substantial heterogeneity across 12 chosen event types/settings, with the highest transmission (25-35%) in co-living situations including households, nursing homes, and other congregate housing. Among index cases, 67% reported zero secondary cases and only 3% (287) infected >5 secondary cases ("superspreaders"). Index case demographic data were limited, with only 55% of individuals reporting age, sex, symptoms, real-time polymerase chain reaction (PCR) cycle threshold values, or total contacts. With the data available, we identified a higher percentage of superspreaders among symptomatic individuals, individuals aged 49-64 years, and individuals with over 100 total contacts. Addressing gaps in the literature regarding transmission events and contact tracing is needed to properly explain the heterogeneity in transmission and facilitate control efforts for SARS-CoV-2 and other infections.
Collapse
Affiliation(s)
- Clifton D. McKee
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Emma X. Yu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrés Garcia
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jules Jackson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Aybüke Koyuncu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sophie Rose
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew S. Azman
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Katie Lobner
- Welch Medical Library, Johns Hopkins University, Baltimore, MD, USA
| | - Emma Sacks
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Maria D. Van Kerkhove
- Department of Epidemic and Pandemic Preparedness and Prevention, Emergency Preparedness Programme, World Health Organization, Geneva, Switzerland
| | - Emily S. Gurley
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
21
|
Phan T, Ribeiro RM, Edelstein GE, Boucau J, Uddin R, Marino C, Liew MY, Barry M, Choudhary MC, Tien D, Su K, Reynolds Z, Li Y, Sagar S, Vyas TD, Kawano Y, Sparks JA, Hammond SP, Wallace Z, Vyas JM, Li JZ, Siedner MJ, Barczak AK, Lemieux JE, Perelson AS. Modeling suggests SARS-CoV-2 rebound after nirmatrelvir-ritonavir treatment is driven by target cell preservation coupled with incomplete viral clearance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.613000. [PMID: 39345409 PMCID: PMC11429690 DOI: 10.1101/2024.09.13.613000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
In a subset of SARS-CoV-2 infected individuals treated with the oral antiviral nirmatrelvir-ritonavir, the virus rebounds following treatment. The mechanisms driving this rebound are not well understood. We used a mathematical model to describe the longitudinal viral load dynamics of 51 individuals treated with nirmatrelvir-ritonavir, 20 of whom rebounded. Target cell preservation, either by a robust innate immune response or initiation of nirmatrelvir-ritonavir near the time of symptom onset, coupled with incomplete viral clearance, appear to be the main factors leading to viral rebound. Moreover, the occurrence of viral rebound is likely influenced by time of treatment initiation relative to the progression of the infection, with earlier treatments leading to a higher chance of rebound. Finally, our model demonstrates that extending the course of nirmatrelvir-ritonavir treatment, in particular to a 10-day regimen, may greatly diminish the risk for rebound in people with mild-to-moderate COVID-19 and who are at high risk of progression to severe disease. Altogether, our results suggest that in some individuals, a standard 5-day course of nirmatrelvir-ritonavir starting around the time of symptom onset may not completely eliminate the virus. Thus, after treatment ends, the virus can rebound if an effective adaptive immune response has not fully developed. These findings on the role of target cell preservation and incomplete viral clearance also offer a possible explanation for viral rebounds following other antiviral treatments for SARS-CoV-2.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87544, USA
| | - Ruy M. Ribeiro
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87544, USA
| | - Gregory E. Edelstein
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Julie Boucau
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Rockib Uddin
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
| | - Caitlin Marino
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - May Y. Liew
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
| | - Mamadou Barry
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
| | - Manish C. Choudhary
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dessie Tien
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
| | - Karry Su
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
| | - Zahra Reynolds
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
| | - Yijia Li
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
- University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA
| | - Shruti Sagar
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
| | - Tammy D. Vyas
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
| | - Yumeko Kawano
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey A. Sparks
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah P. Hammond
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
| | - Zachary Wallace
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
| | - Jatin M. Vyas
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
| | - Jonathan Z. Li
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mark J. Siedner
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
- Africa Health Research Institute, KwaZulu-Natal, South Africa
| | - Amy K. Barczak
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
| | - Jacob E. Lemieux
- Department of Medicine, Massachusetts General Hospital, Havard Medical School, Boston, MA 02114, USA
- Broad Institute, Cambridge, MA 02142, USA
| | - Alan S. Perelson
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87544, USA
- Santa Fe Institute, Santa Fe, NM 87501, USA
| |
Collapse
|
22
|
Braeye T, Proesmans K, Van Cauteren D, Brondeel R, Hens N, Vermeiren E, Hammami N, Rosas A, Taame A, André E, Cuypers L. Personal characteristics and transmission dynamics associated with SARS-CoV-2 semi-quantitative PCR test results: an observational study from Belgium, 2021-2022. Front Public Health 2024; 12:1429021. [PMID: 39319296 PMCID: PMC11420023 DOI: 10.3389/fpubh.2024.1429021] [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: 05/07/2024] [Accepted: 08/20/2024] [Indexed: 09/26/2024] Open
Abstract
Introduction Following harmonization efforts by the Belgian National Reference Center for SARS-CoV-2, semi-quantitative PCR test (SQ-PCR) results, used as a proxy for viral load, were routinely collected after performing RT-qPCR tests. Methods We investigated both the personal characteristics associated with SQ-PCR results and the transmission dynamics involving these results. We used person-level laboratory test data and contact tracing data collected in Belgium from March 2021 to February 2022. Personal characteristics (age, sex, vaccination, and laboratory-confirmed prior infection) and disease stage by date of symptom onset were analyzed in relation to SQ-PCR results using logistic regression. Vaccine effectiveness (VE) against a high viral load (≥107 copies/mL) was estimated from the adjusted probabilities. Contact tracing involves the mandatory testing of high-risk exposure contacts (HREC) after contact with an index case. Odds ratios for test positivity and high viral load in HREC were calculated based on the SQ-PCR result of the index case using logistic regression models adjusted for age, sex, immunity status (vaccination, laboratory-confirmed prior infection), variant (Alpha, Delta, Omicron), calendar time, and contact tracing covariates. Results We included 909,157 SQ-PCR results of COVID-19 cases, 379,640 PCR results from index cases, and 72,052 SQ-PCR results of HREC. High viral load was observed more frequently among recent cases, symptomatic cases, cases over 25 years of age, and those not recently vaccinated (>90 days). The vaccine effectiveness (VE) of the primary schedule in the first 30 days after vaccination was estimated at 47.3% (95%CI 40.8-53.2) during the Delta variant period. A high viral load in index cases was associated with an increased test positivity in HREC (OR 2.7, 95%CI 2.62-2.79) and, among those testing positive, an increased likelihood of a high viral load (OR 2.84, 95%CI 2.53-3.19).
Collapse
Affiliation(s)
- Toon Braeye
- Epidemiology of Infectious Diseases, Sciensano, Brussels, Belgium
- I-BioStat, Data Science Institute, Hasselt University, Hasselt, Belgium
| | - Kristiaan Proesmans
- Faculty of Pharmaceutical Sciences, Department of Bio-analysis, Ghent University, Ghent, Belgium
| | | | - Ruben Brondeel
- Epidemiology of Infectious Diseases, Sciensano, Brussels, Belgium
| | - Niel Hens
- I-BioStat, Data Science Institute, Hasselt University, Hasselt, Belgium
- Centre for Health Economic Research and Modelling Infectious Diseases, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Elias Vermeiren
- Epidemiology of Infectious Diseases, Sciensano, Brussels, Belgium
| | - Naïma Hammami
- Department of Care, Infection Prevention and Control, Flemish Community, Brussels, Belgium
| | - Angel Rosas
- Direction Surveillance des Maladies Infectieuses, Agence out une Vie de Qualité (AVIQ), Charleroi, Belgium
| | - Adrae Taame
- Cellule de médecine préventive- Direction santé et aide aux personnes – Vivalis/Cocom, Brussels, Belgium
| | - Emmanuel André
- Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium
| | - Lize Cuypers
- Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium
| |
Collapse
|
23
|
Hawley DM, Pérez-Umphrey AA, Adelman JS, Fleming-Davies AE, Garrett-Larsen J, Geary SJ, Childs LM, Langwig KE. Prior exposure to pathogens augments host heterogeneity in susceptibility and has key epidemiological consequences. PLoS Pathog 2024; 20:e1012092. [PMID: 39231171 PMCID: PMC11404847 DOI: 10.1371/journal.ppat.1012092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/16/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024] Open
Abstract
Pathogen epidemics are key threats to human and wildlife health. Across systems, host protection from pathogens following initial exposure is often incomplete, resulting in recurrent epidemics through partially-immune hosts. Variation in population-level protection has important consequences for epidemic dynamics, but how acquired protection influences inter-individual heterogeneity in susceptibility and its epidemiological consequences remains understudied. We experimentally investigated whether prior exposure (none, low-dose, or high-dose) to a bacterial pathogen alters host heterogeneity in susceptibility among songbirds. Hosts with no prior pathogen exposure had little variation in protection, but heterogeneity in susceptibility was significantly augmented by prior pathogen exposure, with the highest variability detected in hosts given high-dose prior exposure. An epidemiological model parameterized with experimental data found that heterogeneity in susceptibility from prior exposure more than halved epidemic sizes compared with a homogeneous population with identical mean protection. However, because infection-induced mortality was also greatly reduced in hosts with prior pathogen exposure, reductions in epidemic size were smaller than expected in hosts with prior exposure. These results highlight the importance of variable protection from prior exposure and/or vaccination in driving population-level heterogeneity and epidemiological dynamics.
Collapse
Affiliation(s)
- Dana M Hawley
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virgina, United States of America
| | - Anna A Pérez-Umphrey
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virgina, United States of America
| | - James S Adelman
- Department of Biological Sciences, University of Memphis, Memphis, Tennessee, United States of America
| | | | - Jesse Garrett-Larsen
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virgina, United States of America
| | - Steven J Geary
- Department of Pathobiology & Veterinary Science, University of Connecticut, Storrs, Connecticut, United States of America
| | - Lauren M Childs
- Department of Mathematics and Virginia Tech Center for the Mathematics of Biosystems, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Kate E Langwig
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virgina, United States of America
| |
Collapse
|
24
|
Claas AM, Lee M, Huang PH, Knutson CG, Bullara D, Schoeberl B, Gaudet S. Viral Kinetics Model of SARS-CoV-2 Infection Informs Drug Discovery, Clinical Dose, and Regimen Selection. Clin Pharmacol Ther 2024; 116:757-769. [PMID: 38676291 DOI: 10.1002/cpt.3267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/18/2024] [Indexed: 04/28/2024]
Abstract
Quantitative systems pharmacology (QSP) has been an important tool to project safety and efficacy of novel or repurposed therapies for the SARS-CoV-2 virus. Here, we present a QSP modeling framework to predict response to antiviral therapeutics with three mechanisms of action (MoA): cell entry inhibitors, anti-replicatives, and neutralizing biologics. We parameterized three distinct model structures describing virus-host interaction by fitting to published viral kinetics data of untreated COVID-19 patients. The models were used to test theoretical behaviors and map therapeutic design criteria of the different MoAs, identifying the most rapid and robust antiviral activity from neutralizing biologic and anti-replicative MoAs. We found good agreement between model predictions and clinical viral load reduction observed with anti-replicative nirmatrelvir/ritonavir (Paxlovid®) and neutralizing biologics bamlanivimab and casirivimab/imdevimab (REGEN-COV®), building confidence in the modeling framework to inform a dose selection. Finally, the model was applied to predict antiviral response with ensovibep, a novel DARPin therapeutic designed as a neutralizing biologic. We developed a new in silico measure of antiviral activity, area under the curve (AUC) of free spike protein concentration, as a metric with larger dynamic range than viral load reduction. By benchmarking to bamlanivimab predictions, we justified dose levels of 75, 225, and 600 mg ensovibep to be administered intravenously in a Phase 2 clinical investigation. Upon trial completion, we found model predictions to be in good agreement with the observed patient data. These results demonstrate the utility of this modeling framework to guide the development of novel antiviral therapeutics.
Collapse
Affiliation(s)
- Allison M Claas
- Biomedical Research, Novartis, Cambridge, Massachusetts, USA
| | - Meelim Lee
- Biomedical Research, Novartis, Cambridge, Massachusetts, USA
| | - Pai-Hsi Huang
- Biomedical Research, Novartis, East Hanover, New Jersey, USA
| | | | | | | | - Suzanne Gaudet
- Biomedical Research, Novartis, Cambridge, Massachusetts, USA
| |
Collapse
|
25
|
Wongnak P, Schilling WHK, Jittamala P, Boyd S, Luvira V, Siripoon T, Ngamprasertchai T, Batty EM, Singh S, Kouhathong J, Pagornrat W, Khanthagan P, Hanboonkunupakarn B, Poovorawan K, Mayxay M, Chotivanich K, Imwong M, Pukrittayakamee S, Ashley EA, Dondorp AM, Day NPJ, Teixeira MM, Piyaphanee W, Phumratanaprapin W, White NJ, Watson JA. Temporal changes in SARS-CoV-2 clearance kinetics and the optimal design of antiviral pharmacodynamic studies: an individual patient data meta-analysis of a randomised, controlled, adaptive platform study (PLATCOV). THE LANCET. INFECTIOUS DISEASES 2024; 24:953-963. [PMID: 38677300 DOI: 10.1016/s1473-3099(24)00183-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Effective antiviral drugs prevent hospitalisation and death from COVID-19. Antiviral efficacy can be efficiently assessed in vivo by measuring rates of SARS-CoV-2 clearance estimated from serial viral genome densities quantitated in nasopharyngeal or oropharyngeal swab eluates. We conducted an individual patient data meta-analysis of unblinded arms in the PLATCOV platform trial to characterise changes in viral clearance kinetics and infer optimal design and interpretation of antiviral pharmacometric evaluations. METHODS Serial viral density data were analysed from symptomatic, previously healthy, adult patients (within 4 days of symptom onset) enrolled in a large multicentre, randomised, adaptive, pharmacodynamic, platform trial (PLATCOV) comparing antiviral interventions for SARS-CoV-2. Viral clearance rates over 1 week were estimated under a hierarchical Bayesian linear model with B-splines used to characterise temporal changes in enrolment viral densities and clearance rates. Bootstrap re-sampling was used to assess the optimal duration of follow-up for pharmacometric assessment, where optimal was defined as maximising the expected Z score when comparing effective antivirals with no treatment. PLATCOV is registered at ClinicalTrials.gov, NCT05041907. FINDINGS Between Sept 29, 2021, and Oct 20, 2023, 1262 patients were randomly assigned in the PLATCOV trial. Unblinded data were available from 800 patients (who provided 16 818 oropharyngeal viral quantitative PCR [qPCR] measurements), of whom 504 (63%) were female. 783 (98%) patients had received at least one vaccine dose and 703 (88%) were fully vaccinated. SARS-CoV-2 viral clearance was biphasic (bi-exponential). The first phase (α) was accelerated by effective interventions. For all the effective interventions studied, maximum discriminative power (maximum expected Z score) was obtained when evaluating serial data from the first 5 days after enrolment. Over the 2-year period studied, median viral clearance half-lives estimated over 7 days shortened from 16·6 h (IQR 15·3 to 18·2) in September, 2021, to 9·2 h (8·0 to 10·6) in October, 2023, in patients receiving no antiviral drugs, equivalent to a relative reduction of 44% (95% credible interval [CrI] 19 to 64). A parallel reduction in viral clearance half-lives over time was observed in patients receiving antiviral drugs. For example, in the 158 patients assigned to ritonavir-boosted nirmatrelvir (3380 qPCR measurements), the median viral clearance half-life reduced from 6·4 h (IQR 5·7 to 7·3) in June, 2022, to 4·8 h (4·2 to 5·5) in October, 2023, a relative reduction of 26% (95% CrI -4 to 42). INTERPRETATION SARS-CoV-2 viral clearance kinetics in symptomatic, vaccinated individuals accelerated substantially over 2 years of the pandemic, necessitating a change to how new SARS-CoV-2 antivirals are compared (ie, shortening the period of pharmacodynamic assessment). As of writing (October, 2023), antiviral efficacy in COVID-19 can be efficiently assessed in vivo using serial qPCRs from duplicate oropharyngeal swab eluates taken daily for 5 days after drug administration. FUNDING Wellcome Trust.
Collapse
Affiliation(s)
- Phrutsamon Wongnak
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - William H K Schilling
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Podjanee Jittamala
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Department of Tropical Hygiene, Mahidol University, Bangkok, Thailand
| | - Simon Boyd
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Viravarn Luvira
- Department of Clinical Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Tanaya Siripoon
- Department of Clinical Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Elizabeth M Batty
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Shivani Singh
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Jindarat Kouhathong
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Watcharee Pagornrat
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Patpannee Khanthagan
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Borimas Hanboonkunupakarn
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Department of Clinical Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kittiyod Poovorawan
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Department of Clinical Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Mayfong Mayxay
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK; Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Laos; Institute for Research and Education Development, University of Health Sciences, Vientiane, Laos
| | - Kesinee Chotivanich
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Department of Clinical Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Mallika Imwong
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Department of Molecular Tropical Medicine and Genetics, Mahidol University, Bangkok, Thailand
| | - Sasithon Pukrittayakamee
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Department of Clinical Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Elizabeth A Ashley
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK; Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Laos
| | - Arjen M Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Nicholas P J Day
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Mauro M Teixeira
- Clinical Research Unit, Center for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Brazil
| | | | | | - Nicholas J White
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK.
| | - James A Watson
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK; Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam.
| |
Collapse
|
26
|
Hawley DM, Pérez-Umphrey AA, Adelman JS, Fleming-Davies AE, Garrett-Larsen J, Geary SJ, Childs LM, Langwig KE. Prior exposure to pathogens augments host heterogeneity in susceptibility and has key epidemiological consequences. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583455. [PMID: 38496428 PMCID: PMC10942282 DOI: 10.1101/2024.03.05.583455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Pathogen epidemics are key threats to human and wildlife health. Across systems, host protection from pathogens following initial exposure is often incomplete, resulting in recurrent epidemics through partially-immune hosts. Variation in population-level protection has important consequences for epidemic dynamics, but how acquired protection influences inter-individual heterogeneity in susceptibility and its epidemiological consequences remains understudied. We experimentally investigated whether prior exposure (none, low-dose, or high-dose) to a bacterial pathogen alters host heterogeneity in susceptibility among songbirds. Hosts with no prior pathogen exposure had little variation in protection, but heterogeneity in susceptibility was significantly augmented by prior pathogen exposure, with the highest variability detected in hosts given high-dose prior exposure. An epidemiological model parameterized with experimental data found that heterogeneity in susceptibility from prior exposure more than halved epidemic sizes compared with a homogeneous population with identical mean protection. However, because infection-induced mortality was also greatly reduced in hosts with prior pathogen exposure, reductions in epidemic size were smaller than expected in hosts with prior exposure. These results highlight the importance of variable protection from prior exposure and/or vaccination in driving population-level heterogeneity and epidemiological dynamics.
Collapse
Affiliation(s)
- Dana M. Hawley
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | | | - James S. Adelman
- Department of Biological Sciences, University of Memphis, Memphis, TN, USA
| | | | | | - Steven J. Geary
- Department of Pathobiology & Veterinary Science, University of Connecticut, Storrs, CT, USA
| | - Lauren M. Childs
- Department of Mathematics and Virginia Tech Center for Mathematics of Biosystems, Virginia Tech, Blacksburg, VA, USA
| | - Kate E. Langwig
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
27
|
Kim AE, Bennett JC, Luiten K, O'Hanlon JA, Wolf CR, Magedson A, Han PD, Acker Z, Regelbrugge L, McCaffrey KM, Stone J, Reinhart D, Capodanno BJ, Morse SS, Bedford T, Englund JA, Boeckh M, Starita LM, Uyeki TM, Carone M, Weil A, Chu HY. Comparative Diagnostic Utility of SARS-CoV-2 Rapid Antigen and Molecular Testing in a Community Setting. J Infect Dis 2024; 230:363-373. [PMID: 38531685 DOI: 10.1093/infdis/jiae150] [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: 12/08/2023] [Revised: 03/12/2024] [Accepted: 03/25/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND SARS-CoV-2 antigen-detection rapid diagnostic tests (Ag-RDTs) have become widely utilized but longitudinal characterization of their community-based performance remains incompletely understood. METHODS This prospective longitudinal study at a large public university in Seattle, WA utilized remote enrollment, online surveys, and self-collected nasal swab specimens to evaluate Ag-RDT performance against real-time reverse transcription polymerase chain reaction (rRT-PCR) in the context of SARS-CoV-2 Omicron. Ag-RDT sensitivity and specificity within 1 day of rRT-PCR were evaluated by symptom status throughout the illness episode and Orf1b cycle threshold (Ct). RESULTS From February to December 2022, 5757 participants reported 17 572 Ag-RDT results and completed 12 674 rRT-PCR tests, of which 995 (7.9%) were rRT-PCR positive. Overall sensitivity and specificity were 53.0% (95% confidence interval [CI], 49.6%-56.4%) and 98.8% (95% CI, 98.5%-99.0%), respectively. Sensitivity was comparatively higher for Ag-RDTs used 1 day after rRT-PCR (69.0%), 4-7 days after symptom onset (70.1%), and Orf1b Ct ≤20 (82.7%). Serial Ag-RDT sensitivity increased with repeat testing ≥2 (68.5%) and ≥4 (75.8%) days after an initial Ag-RDT-negative result. CONCLUSIONS Ag-RDT performance varied by clinical characteristics and temporal testing patterns. Our findings support recommendations for serial testing following an initial Ag-RDT-negative result, especially among recently symptomatic persons or those at high risk for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ashley E Kim
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Julia C Bennett
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Kyle Luiten
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Jessica A O'Hanlon
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Caitlin R Wolf
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Ariana Magedson
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Peter D Han
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Zack Acker
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Lani Regelbrugge
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | | | - Jeremey Stone
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - David Reinhart
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Benjamin J Capodanno
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Stephen S Morse
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Trevor Bedford
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Howard Hughes Medical Institute, Seattle, Washington, USA
| | - Janet A Englund
- Seattle Children's Research Institute, Seattle, Washington, USA
| | - Michael Boeckh
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Lea M Starita
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Timothy M Uyeki
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Marco Carone
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Ana Weil
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Helen Y Chu
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
28
|
Salzano L, Narayanan N, Tobik ER, Akbarzada S, Wu Y, Megiel S, Choate B, Wyllie AL. Diagnostic testing preferences can help inform future public health response efforts: Global insights from an international survey. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003547. [PMID: 39078819 PMCID: PMC11288416 DOI: 10.1371/journal.pgph.0003547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 07/08/2024] [Indexed: 08/02/2024]
Abstract
Public perception regarding diagnostic sample types as well as personal experiences can influence willingness to test. As such, public preferences for specific sample type(s) should be used to inform diagnostic and surveillance testing programs to improve public health response efforts. To understand where preferences lie, we conducted an international survey regarding the sample types used for SARS-CoV-2 tests. A Qualtrics survey regarding SARS-CoV-2 testing preferences was distributed via social media and email. The survey collected preferences regarding sample methods and key demographic data. Python was used to analyze survey responses. From March 30th to June 15th, 2022, 2,094 responses were collected from 125 countries. Participants were 55% female and predominantly aged 25-34 years (27%). Education and employment were skewed: 51% had graduate degrees, 26% had bachelor's degrees, 27% were scientists/researchers, and 29% were healthcare workers. By rank sum analysis, the most preferred sample type globally was the oral swab, followed by saliva, with parents/guardians preferring saliva-based testing for children. Respondents indicated a higher degree of trust in PCR testing (84%) vs. rapid antigen testing (36%). Preferences for self- or healthcare worker-collected sampling varied across regions. This international survey identified a preference for oral swabs and saliva when testing for SARS-CoV-2. Notably, respondents indicated that if they could be assured that all sample types performed equally, then saliva was preferred. Overall, survey responses reflected the region-specific testing experiences during the COVID-19. Public preferences should be considered when designing future response efforts to increase utilization, with oral sample types (either swabs or saliva) providing a practical option for large-scale, accessible diagnostic testing.
Collapse
Affiliation(s)
- Leah Salzano
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Nithya Narayanan
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Emily R. Tobik
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Sumaira Akbarzada
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Yanjun Wu
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Sarah Megiel
- Department of Health Policy and Management, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Brittany Choate
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
- SalivaDirect, Inc., New Haven, Connecticut, United States of America
| | - Anne L. Wyllie
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| |
Collapse
|
29
|
Samsunder N, Sivro A, Hassan-Moosa R, Lewis L, Kara Z, Baxter C, Karim QA, Karim SA, Kharsany ABM, Naidoo K, Ngcapu S. Evaluating diagnostic accuracy of an RT-PCR test for the detection of SARS-CoV-2 in saliva. Diagn Progn Res 2024; 8:9. [PMID: 39044271 PMCID: PMC11267770 DOI: 10.1186/s41512-024-00176-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 07/01/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Saliva has been proposed as a potential more convenient, cost-effective, and easier sample for diagnosing SARS-CoV-2 infections, but there is limited knowledge of the impact of saliva volumes and stages of infection on its sensitivity and specificity. METHODS In this study, we assessed the performance of SARS-CoV-2 testing in 171 saliva samples from 52 mostly mildly symptomatic patients (aged 18 to 70 years) with a positive reference standard result at screening. The samples were collected at different volumes (50, 100, 300, and 500 µl of saliva) and at different stages of the disease (at enrollment, day 7, 14, and 28 post SARS-CoV-2 diagnosis). Imperfect nasopharyngeal (NP) swab nucleic acid amplification testing was used as a reference. We used a logistic regression with generalized estimating equations to estimate sensitivity, specificity, PPV, and NPV, accounting for the correlation between repeated observations. RESULTS The sensitivity and specificity values were consistent across saliva volumes. The sensitivity of saliva samples ranged from 70.2% (95% CI, 49.3-85.0%) for 100 μl to 81.0% (95% CI, 51.9-94.4%) for 300 μl of saliva collected. The specificity values ranged between 75.8% (95% CI, 55.0-88.9%) for 50 μl and 78.8% (95% CI, 63.2-88.9%) for 100 μl saliva compared to NP swab samples. The overall percentage of positive results in NP swabs and saliva specimens remained comparable throughout the study visits. We observed no significant difference in cycle number values between saliva and NP swab specimens, irrespective of saliva volume tested. CONCLUSIONS The saliva collection offers a promising approach for population-based testing.
Collapse
Affiliation(s)
- Natasha Samsunder
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
| | - Aida Sivro
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
- JC Wilt Infectious Disease Research Centre, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Razia Hassan-Moosa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
- SAMRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Lara Lewis
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
| | - Zahra Kara
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
| | - Cheryl Baxter
- Centre for Epidemic Response and Innovation (CERI), Stellenbosch University, Stellenbosch, South Africa
| | - Quarraisha Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Salim Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Ayesha B M Kharsany
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
- SAMRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Sinaye Ngcapu
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa.
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
30
|
Esmaeili S, Owens K, Wagoner J, Polyak SJ, White JM, Schiffer JT. A unifying model to explain frequent SARS-CoV-2 rebound after nirmatrelvir treatment and limited prophylactic efficacy. Nat Commun 2024; 15:5478. [PMID: 38942778 PMCID: PMC11213957 DOI: 10.1038/s41467-024-49458-9] [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: 02/06/2024] [Accepted: 06/04/2024] [Indexed: 06/30/2024] Open
Abstract
In a pivotal trial (EPIC-HR), a 5-day course of oral ritonavir-boosted nirmatrelvir, given early during symptomatic SARS-CoV-2 infection (within three days of symptoms onset), decreased hospitalization and death by 89.1% and nasal viral load by 0.87 log relative to placebo in high-risk individuals. Yet, nirmatrelvir/ritonavir failed as post-exposure prophylaxis in a trial, and frequent viral rebound has been observed in subsequent cohorts. We develop a mathematical model capturing viral-immune dynamics and nirmatrelvir pharmacokinetics that recapitulates viral loads from this and another clinical trial (PLATCOV). Our results suggest that nirmatrelvir's in vivo potency is significantly lower than in vitro assays predict. According to our model, a maximally potent agent would reduce the viral load by approximately 3.5 logs relative to placebo at 5 days. The model identifies that earlier initiation and shorter treatment duration are key predictors of post-treatment rebound. Extension of treatment to 10 days for Omicron variant infection in vaccinated individuals, rather than increasing dose or dosing frequency, is predicted to lower the incidence of viral rebound significantly.
Collapse
Affiliation(s)
- Shadisadat Esmaeili
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Katherine Owens
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jessica Wagoner
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Stephen J Polyak
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Judith M White
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - Joshua T Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
31
|
Middleton C, Larremore DB. Modeling the transmission mitigation impact of testing for infectious diseases. SCIENCE ADVANCES 2024; 10:eadk5108. [PMID: 38875334 PMCID: PMC11177932 DOI: 10.1126/sciadv.adk5108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 05/10/2024] [Indexed: 06/16/2024]
Abstract
A fundamental question of any program focused on the testing and timely diagnosis of a communicable disease is its effectiveness in reducing transmission. Here, we introduce testing effectiveness (TE)-the fraction by which testing and post-diagnosis isolation reduce transmission at the population scale-and a model that incorporates test specifications and usage, within-host pathogen dynamics, and human behaviors to estimate TE. Using TE to guide recommendations, we show that today's rapid diagnostics should be used immediately upon symptom onset to control influenza A and respiratory syncytial virus but delayed by up to two days to control omicron-era severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Furthermore, while rapid tests are superior to reverse transcription quantitative polymerase chain reaction (RT-qPCR) to control founder-strain SARS-CoV-2, omicron-era changes in viral kinetics and rapid test sensitivity cause a reversal, with higher TE for RT-qPCR despite longer turnaround times. Last, we illustrate the model's flexibility by quantifying trade-offs in the use of post-diagnosis testing to shorten isolation times.
Collapse
Affiliation(s)
- Casey Middleton
- Department of Computer Science, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Daniel B. Larremore
- Department of Computer Science, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
- Santa Fe Institute, Santa Fe, NM, USA
| |
Collapse
|
32
|
Herbert C, Wang B, Lin H, Yan Y, Hafer N, Pretz C, Stamegna P, Wright C, Suvarna T, Harman E, Schrader S, Nowak C, Kheterpal V, Orvek E, Wong S, Zai A, Barton B, Gerber BS, Lemon SC, Filippaios A, Gibson L, Greene S, Colubri A, Achenbach C, Murphy R, Heetderks W, Manabe YC, O’Connor L, Fahey N, Luzuriaga K, Broach J, Roth K, McManus DD, Soni A. Performance of and Severe Acute Respiratory Syndrome Coronavirus 2 Diagnostics Based on Symptom Onset and Close Contact Exposure: An Analysis From the Test Us at Home Prospective Cohort Study. Open Forum Infect Dis 2024; 11:ofae304. [PMID: 38911947 PMCID: PMC11191649 DOI: 10.1093/ofid/ofae304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/29/2024] [Indexed: 06/25/2024] Open
Abstract
Background Understanding changes in diagnostic performance after symptom onset and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exposure within different populations is crucial to guide the use of diagnostics for SARS-CoV-2. Methods The Test Us at Home study was a longitudinal cohort study that enrolled individuals across the United States between October 2021 and February 2022. Participants performed paired antigen-detection rapid diagnostic tests (Ag-RDTs) and reverse-transcriptase polymerase chain reaction (RT-PCR) tests at home every 48 hours for 15 days and self-reported symptoms and known coronavirus disease 2019 exposures immediately before testing. The percent positivity for Ag-RDTs and RT-PCR tests was calculated each day after symptom onset and exposure and stratified by vaccination status, variant, age category, and sex. Results The highest percent positivity occurred 2 days after symptom onset (RT-PCR, 91.2%; Ag-RDT, 71.1%) and 6 days after exposure (RT-PCR, 91.8%; Ag-RDT, 86.2%). RT-PCR and Ag-RDT performance did not differ by vaccination status, variant, age category, or sex. The percent positivity for Ag-RDTs was lower among exposed, asymptomatic than among symptomatic individuals (37.5% (95% confidence interval [CI], 13.7%-69.4%) vs 90.3% (75.1%-96.7%). Cumulatively, Ag-RDTs detected 84.9% (95% CI, 78.2%-89.8%) of infections within 4 days of symptom onset. For exposed participants, Ag-RDTs detected 94.0% (95% CI, 86.7%-97.4%) of RT-PCR-confirmed infections within 6 days of exposure. Conclusions The percent positivity for Ag-RDTs and RT-PCR tests was highest 2 days after symptom onset and 6 days after exposure, and performance increased with serial testing. The percent positivity of Ag-RDTs was lowest among asymptomatic individuals but did not differ by sex, variant, vaccination status, or age category.
Collapse
Affiliation(s)
- Carly Herbert
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- University of Massachusetts Center for Clinical and Translational Science, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Biqi Wang
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Division of Health System Science, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Honghuang Lin
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Division of Health System Science, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Yi Yan
- Division of Microbiology, OHT7 Office of Product Evaluation and Quality, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Nathaniel Hafer
- University of Massachusetts Center for Clinical and Translational Science, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Caitlin Pretz
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Pamela Stamegna
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Colton Wright
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | | | | | | | | | | | - Elizabeth Orvek
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Steven Wong
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Adrian Zai
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Bruce Barton
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Ben S Gerber
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Stephenie C Lemon
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Andreas Filippaios
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Laura Gibson
- Division of Infectious Disease, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Sharone Greene
- Division of Infectious Disease, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Andres Colubri
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Chad Achenbach
- Division of Infectious Disease, Department of Medicine, Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Robert Murphy
- Division of Infectious Disease, Department of Medicine, Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - William Heetderks
- National Institute of Biomedical Imaging and Bioengineering, NIH, via contract with Kelly Services, Bethesda, Maryland, USA
| | - Yukari C Manabe
- Division of Infectious Disease, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Laurel O’Connor
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Nisha Fahey
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Katherine Luzuriaga
- University of Massachusetts Center for Clinical and Translational Science, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - John Broach
- University of Massachusetts Center for Clinical and Translational Science, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Kristian Roth
- Division of Microbiology, OHT7 Office of Product Evaluation and Quality, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - David D McManus
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Division of Health System Science, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Division of Cardiology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Apurv Soni
- Program in Digital Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Division of Health System Science, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
33
|
Schuh L, Markov PV, Veliov VM, Stilianakis NI. A mathematical model for the within-host (re)infection dynamics of SARS-CoV-2. Math Biosci 2024; 371:109178. [PMID: 38490360 PMCID: PMC11636724 DOI: 10.1016/j.mbs.2024.109178] [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: 12/12/2023] [Revised: 02/08/2024] [Accepted: 03/11/2024] [Indexed: 03/17/2024]
Abstract
Interactions between SARS-CoV-2 and the immune system during infection are complex. However, understanding the within-host SARS-CoV-2 dynamics is of enormous importance for clinical and public health outcomes. Current mathematical models focus on describing the within-host SARS-CoV-2 dynamics during the acute infection phase. Thereby they ignore important long-term post-acute infection effects. We present a mathematical model, which not only describes the SARS-CoV-2 infection dynamics during the acute infection phase, but extends current approaches by also recapitulating clinically observed long-term post-acute infection effects, such as the recovery of the number of susceptible epithelial cells to an initial pre-infection homeostatic level, a permanent and full clearance of the infection within the individual, immune waning, and the formation of long-term immune capacity levels after infection. Finally, we used our model and its description of the long-term post-acute infection dynamics to explore reinfection scenarios differentiating between distinct variant-specific properties of the reinfecting virus. Together, the model's ability to describe not only the acute but also the long-term post-acute infection dynamics provides a more realistic description of key outcomes and allows for its application in clinical and public health scenarios.
Collapse
Affiliation(s)
- Lea Schuh
- Joint Research Centre (JRC), European Commission, Via Enrico Fermi 2749, Ispra, 21027, Italy.
| | - Peter V Markov
- Joint Research Centre (JRC), European Commission, Via Enrico Fermi 2749, Ispra, 21027, Italy; London School of Hygiene & Tropical Medicine, University of London, Keppel Street, London, WC1E 7HT, United Kingdom
| | - Vladimir M Veliov
- Institute of Statistics and Mathematical Methods in Economics, Vienna University of Technology, Wiedner Hauptstraße 8-10, Vienna, 1040, Austria
| | - Nikolaos I Stilianakis
- Joint Research Centre (JRC), European Commission, Via Enrico Fermi 2749, Ispra, 21027, Italy; Department of Biometry and Epidemiology, University of Erlangen-Nuremberg, Waldstraße 6, Erlangen, 91054, Germany.
| |
Collapse
|
34
|
Zhang W, Gai X, Wang B, Duan Z, Zhou Q, Dai L, Yan C, Wu C, Fan J, Wang P, Yang P, Bao F, Jing H, Cai C, Song C, Ma Y, Sun Y. A robust web-based tool to predict viral shedding in patients with Omicron SARS-CoV-2 variants. ERJ Open Res 2024; 10:00939-2023. [PMID: 38779041 PMCID: PMC11111115 DOI: 10.1183/23120541.00939-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 02/17/2024] [Indexed: 05/25/2024] Open
Abstract
Background Data on viral kinetics and variants affecting the duration of viral shedding were limited. Our objective was to determine viral shedding in distinct severe acute respiratory syndrome coronavirus 2 variants, including Omicron BA.4/5 and BF.7, and to identify the relevant influencing factors. Methods We carried out a longitudinal cohort study at Beijing Xiaotangshan Fangcang shelter hospital from May to June 2022 (Omicron BA.4/5) and from November to December 2022 (Omicron BF.7). Nucleocapsid protein (N) and open reading frame (ORF) genes were considered as the target genes of the reverse transcription PCR. The daily results of cycle threshold (CT), including lowest ORF1ab-CT values for days 1-3 post-hospitalisation and lowest N-CT values for days 1-3 post-hospitalisation (CT3minN) and demographic and clinical characteristics were collected. Results 1433 patients with coronavirus disease 2019 (COVID-19) were recruited from the Fangcang shelter hospital, in which 278 patients were diagnosed with Omicron BA.4/5 and 1155 patients with Omicron BF.7. Patients with BF.7 infection showed a longer duration of viral shedding. The duration of viral shedding was associated with the variants age, alcohol use, the severity of COVID-19 and CT3minN. Moreover, the nomogram had excellent accuracy in predicting viral shedding. Conclusions Our results indicated that patients with Omicron BF.7 had a longer period of contagiousness than those with BA.4/5. The duration of viral shedding was affected by a variety of factors and the nomogram may become an applicable clinical instrument to predict viral shedding. Furthermore, we developed a new COVID-19 viral shedding predicting model that can accurately predict the duration of viral shedding for COVID-19, and created a user-friendly website to apply this prediction model (https://puh3.shinyapps.io/CVSP_Model/).
Collapse
Affiliation(s)
- Weilong Zhang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
- W. Zhang, X. Gai and B. Wang contributed equally to this article as co-first authors
| | - Xiaoyan Gai
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, and Center for Chronic Airway Diseases, Peking University Health Science Center, Peking University, Beijing, China
- W. Zhang, X. Gai and B. Wang contributed equally to this article as co-first authors
| | - Ben Wang
- Orthopedics Department, Peking University Third Hospital, Beijing, China
- W. Zhang, X. Gai and B. Wang contributed equally to this article as co-first authors
| | - Zhonghui Duan
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Qingtao Zhou
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, and Center for Chronic Airway Diseases, Peking University Health Science Center, Peking University, Beijing, China
| | - Lili Dai
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Changjian Yan
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Chaoling Wu
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Jiarun Fan
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Ping Wang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Ping Yang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Fang Bao
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Hongmei Jing
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Chao Cai
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Chunli Song
- Orthopedics Department, Peking University Third Hospital, Beijing, China
| | - Yingmin Ma
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Y. Ma and Y. Sun contributed equally to this article as lead authors and supervised the work
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, and Center for Chronic Airway Diseases, Peking University Health Science Center, Peking University, Beijing, China
- Y. Ma and Y. Sun contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
35
|
Owens K, Esmaeili S, Schiffer JT. Heterogeneous SARS-CoV-2 kinetics due to variable timing and intensity of immune responses. JCI Insight 2024; 9:e176286. [PMID: 38573774 PMCID: PMC11141931 DOI: 10.1172/jci.insight.176286] [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: 09/29/2023] [Accepted: 03/27/2024] [Indexed: 04/06/2024] Open
Abstract
The viral kinetics of documented SARS-CoV-2 infections exhibit a high degree of interindividual variability. We identified 6 distinct viral shedding patterns, which differed according to peak viral load, duration, expansion rate, and clearance rate, by clustering data from 768 infections in the National Basketball Association cohort. Omicron variant infections in previously vaccinated individuals generally led to lower cumulative shedding levels of SARS-CoV-2 than other scenarios. We then developed a mechanistic mathematical model that recapitulated 1,510 observed viral trajectories, including viral rebound and cases of reinfection. Lower peak viral loads were explained by a more rapid and sustained transition of susceptible cells to a refractory state during infection as well as by an earlier and more potent late, cytolytic immune response. Our results suggest that viral elimination occurs more rapidly during Omicron infection, following vaccination, and following reinfection due to enhanced innate and acquired immune responses. Because viral load has been linked with COVID-19 severity and transmission risk, our model provides a framework for understanding the wide range of observed SARS-CoV-2 infection outcomes.
Collapse
Affiliation(s)
- Katherine Owens
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Shadisadat Esmaeili
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Joshua T. Schiffer
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
- University of Washington, Department of Medicine, Seattle, Washington, USA
| |
Collapse
|
36
|
Snedden CE, Lloyd-Smith JO. Predicting the presence of infectious virus from PCR data: A meta-analysis of SARS-CoV-2 in non-human primates. PLoS Pathog 2024; 20:e1012171. [PMID: 38683864 PMCID: PMC11081500 DOI: 10.1371/journal.ppat.1012171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 05/09/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Researchers and clinicians often rely on molecular assays like PCR to identify and monitor viral infections, instead of the resource-prohibitive gold standard of viral culture. However, it remains unclear when (if ever) PCR measurements of viral load are reliable indicators of replicating or infectious virus. The recent popularity of PCR protocols targeting subgenomic RNA for SARS-CoV-2 has caused further confusion, as the relationships between subgenomic RNA and standard total RNA assays are incompletely characterized and opinions differ on which RNA type better predicts culture outcomes. Here, we explore these issues by comparing total RNA, subgenomic RNA, and viral culture results from 24 studies of SARS-CoV-2 in non-human primates (including 2167 samples from 174 individuals) using custom-developed Bayesian statistical models. On out-of-sample data, our best models predict subgenomic RNA positivity from total RNA data with 91% accuracy, and they predict culture positivity with 85% accuracy. Further analyses of individual time series indicate that many apparent prediction errors may arise from issues with assay sensitivity or sample processing, suggesting true accuracy may be higher than these estimates. Total RNA and subgenomic RNA showed equivalent performance as predictors of culture positivity. Multiple cofactors (including exposure conditions, host traits, and assay protocols) influence culture predictions, yielding insights into biological and methodological sources of variation in assay outcomes-and indicating that no single threshold value applies across study designs. We also show that our model can accurately predict when an individual is no longer infectious, illustrating the potential for future models trained on human data to guide clinical decisions on case isolation. Our work shows that meta-analysis of in vivo data can overcome longstanding challenges arising from limited sample sizes and can yield robust insights beyond those attainable from individual studies. Our analytical pipeline offers a framework to develop similar predictive tools in other virus-host systems, including models trained on human data, which could support laboratory analyses, medical decisions, and public health guidelines.
Collapse
Affiliation(s)
- Celine E. Snedden
- Department of Ecology and Evolutionary Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - James O. Lloyd-Smith
- Department of Ecology and Evolutionary Biology, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Computational Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
37
|
Phan T, Zitzmann C, Chew KW, Smith DM, Daar ES, Wohl DA, Eron JJ, Currier JS, Hughes MD, Choudhary MC, Deo R, Li JZ, Ribeiro RM, Ke R, Perelson AS. Modeling the emergence of viral resistance for SARS-CoV-2 during treatment with an anti-spike monoclonal antibody. PLoS Pathog 2024; 20:e1011680. [PMID: 38635853 PMCID: PMC11060554 DOI: 10.1371/journal.ppat.1011680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 04/30/2024] [Accepted: 03/18/2024] [Indexed: 04/20/2024] Open
Abstract
To mitigate the loss of lives during the COVID-19 pandemic, emergency use authorization was given to several anti-SARS-CoV-2 monoclonal antibody (mAb) therapies for the treatment of mild-to-moderate COVID-19 in patients with a high risk of progressing to severe disease. Monoclonal antibodies used to treat SARS-CoV-2 target the spike protein of the virus and block its ability to enter and infect target cells. Monoclonal antibody therapy can thus accelerate the decline in viral load and lower hospitalization rates among high-risk patients with variants susceptible to mAb therapy. However, viral resistance has been observed, in some cases leading to a transient viral rebound that can be as large as 3-4 orders of magnitude. As mAbs represent a proven treatment choice for SARS-CoV-2 and other viral infections, evaluation of treatment-emergent mAb resistance can help uncover underlying pathobiology of SARS-CoV-2 infection and may also help in the development of the next generation of mAb therapies. Although resistance can be expected, the large rebounds observed are much more difficult to explain. We hypothesize replenishment of target cells is necessary to generate the high transient viral rebound. Thus, we formulated two models with different mechanisms for target cell replenishment (homeostatic proliferation and return from an innate immune response antiviral state) and fit them to data from persons with SARS-CoV-2 treated with a mAb. We showed that both models can explain the emergence of resistant virus associated with high transient viral rebounds. We found that variations in the target cell supply rate and adaptive immunity parameters have a strong impact on the magnitude or observability of the viral rebound associated with the emergence of resistant virus. Both variations in target cell supply rate and adaptive immunity parameters may explain why only some individuals develop observable transient resistant viral rebound. Our study highlights the conditions that can lead to resistance and subsequent viral rebound in mAb treatments during acute infection.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Carolin Zitzmann
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Kara W. Chew
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Davey M. Smith
- Department of Medicine, University of California, San Diego, California, United States of America
| | - Eric S. Daar
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - David A. Wohl
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Joseph J. Eron
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Judith S. Currier
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Michael D. Hughes
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Manish C. Choudhary
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rinki Deo
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jonathan Z. Li
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ruy M. Ribeiro
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Ruian Ke
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Alan S. Perelson
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
- Santa Fe Institute, Santa Fe, New Mexico, United States of America
| | | |
Collapse
|
38
|
Smits HH, Jochems SP. Diverging patterns in innate immunity against respiratory viruses during a lifetime: lessons from the young and the old. Eur Respir Rev 2024; 33:230266. [PMID: 39009407 PMCID: PMC11262623 DOI: 10.1183/16000617.0266-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/16/2024] [Indexed: 07/17/2024] Open
Abstract
Respiratory viral infections frequently lead to severe respiratory disease, particularly in vulnerable populations such as young children, individuals with chronic lung conditions and older adults, resulting in hospitalisation and, in some cases, fatalities. The innate immune system plays a crucial role in monitoring for, and initiating responses to, viruses, maintaining a state of preparedness through the constant expression of antimicrobial defence molecules. Throughout the course of infection, innate immunity remains actively involved, contributing to viral clearance and damage control, with pivotal contributions from airway epithelial cells and resident and newly recruited immune cells. In instances where viral infections persist or are not effectively eliminated, innate immune components prominently contribute to the resulting pathophysiological consequences. Even though both young children and older adults are susceptible to severe respiratory disease caused by various respiratory viruses, the underlying mechanisms may differ significantly. Children face the challenge of developing and maturing their immunity, while older adults contend with issues such as immune senescence and inflammaging. This review aims to compare the innate immune responses in respiratory viral infections across both age groups, identifying common central hubs that could serve as promising targets for innovative therapeutic and preventive strategies, despite the apparent differences in underlying mechanisms.
Collapse
Affiliation(s)
- Hermelijn H Smits
- Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center, Leiden, The Netherlands
| | - Simon P Jochems
- Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
39
|
Townsley H, Gahir J, Russell TW, Greenwood D, Carr EJ, Dyke M, Adams L, Miah M, Clayton B, Smith C, Miranda M, Mears HV, Bailey C, Black JRM, Fowler AS, Crawford M, Wilkinson K, Hutchinson M, Harvey R, O’Reilly N, Kelly G, Goldstone R, Beale R, Papineni P, Corrah T, Gilson R, Caidan S, Nicod J, Gamblin S, Kassiotis G, Libri V, Williams B, Gandhi S, Kucharski AJ, Swanton C, Bauer DLV, Wall EC. COVID-19 in non-hospitalised adults caused by either SARS-CoV-2 sub-variants Omicron BA.1, BA.2, BA.4/5 or Delta associates with similar illness duration, symptom severity and viral kinetics, irrespective of vaccination history. PLoS One 2024; 19:e0294897. [PMID: 38512960 PMCID: PMC10956747 DOI: 10.1371/journal.pone.0294897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 11/11/2023] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND SARS-CoV-2 variant Omicron rapidly evolved over 2022, causing three waves of infection due to sub-variants BA.1, BA.2 and BA.4/5. We sought to characterise symptoms and viral loads over the course of COVID-19 infection with these sub-variants in otherwise-healthy, vaccinated, non-hospitalised adults, and compared data to infections with the preceding Delta variant of concern (VOC). METHODS In a prospective, observational cohort study, healthy vaccinated UK adults who reported a positive polymerase chain reaction (PCR) or lateral flow test, self-swabbed on alternate weekdays until day 10. We compared participant-reported symptoms and viral load trajectories between infections caused by VOCs Delta and Omicron (sub-variants BA.1, BA.2 or BA.4/5), and tested for relationships between vaccine dose, symptoms and PCR cycle threshold (Ct) as a proxy for viral load using Chi-squared (χ2) and Wilcoxon tests. RESULTS 563 infection episodes were reported among 491 participants. Across infection episodes, there was little variation in symptom burden (4 [IQR 3-5] symptoms) and duration (8 [IQR 6-11] days). Whilst symptom profiles differed among infections caused by Delta compared to Omicron sub-variants, symptom profiles were similar between Omicron sub-variants. Anosmia was reported more frequently in Delta infections after 2 doses compared with Omicron sub-variant infections after 3 doses, for example: 42% (25/60) of participants with Delta infection compared to 9% (6/67) with Omicron BA.4/5 (χ2 P < 0.001; OR 7.3 [95% CI 2.7-19.4]). Fever was less common with Delta (20/60 participants; 33%) than Omicron BA.4/5 (39/67; 58%; χ2 P = 0.008; OR 0.4 [CI 0.2-0.7]). Amongst infections with an Omicron sub-variants, symptoms of coryza, fatigue, cough and myalgia predominated. Viral load trajectories and peaks did not differ between Delta, and Omicron, irrespective of symptom severity (including asymptomatic participants), VOC or vaccination status. PCR Ct values were negatively associated with time since vaccination in participants infected with BA.1 (β = -0.05 (CI -0.10-0.01); P = 0.031); however, this trend was not observed in BA.2 or BA.4/5 infections. CONCLUSION Our study emphasises both the changing symptom profile of COVID-19 infections in the Omicron era, and ongoing transmission risk of Omicron sub-variants in vaccinated adults. TRIAL REGISTRATION NCT04750356.
Collapse
Affiliation(s)
- Hermaleigh Townsley
- The Francis Crick Institute, London, United Kingdom
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility, London, United Kingdom
| | - Joshua Gahir
- The Francis Crick Institute, London, United Kingdom
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility, London, United Kingdom
| | - Timothy W. Russell
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | | | - Matala Dyke
- The Francis Crick Institute, London, United Kingdom
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility, London, United Kingdom
| | - Lorin Adams
- Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Murad Miah
- The Francis Crick Institute, London, United Kingdom
| | | | - Callie Smith
- The Francis Crick Institute, London, United Kingdom
| | | | | | - Chris Bailey
- The Francis Crick Institute, London, United Kingdom
| | - James R. M. Black
- The Francis Crick Institute, London, United Kingdom
- University College London, London, United Kingdom
| | | | | | | | | | - Ruth Harvey
- The Francis Crick Institute, London, United Kingdom
- Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | | | - Gavin Kelly
- The Francis Crick Institute, London, United Kingdom
| | | | - Rupert Beale
- The Francis Crick Institute, London, United Kingdom
- University College London, London, United Kingdom
- Genotype-to-Phenotype UK National Virology Consortium (G2P-UK)
| | | | - Tumena Corrah
- London Northwest University Healthcare NHS Trust, London, United Kingdom
| | - Richard Gilson
- Camden and North West London NHS Community Trust, London, United Kingdom
| | - Simon Caidan
- The Francis Crick Institute, London, United Kingdom
| | - Jerome Nicod
- The Francis Crick Institute, London, United Kingdom
| | | | - George Kassiotis
- The Francis Crick Institute, London, United Kingdom
- Department of Infectious Disease, St Mary’s Hospital, Imperial College London, London, United Kingdom
| | - Vincenzo Libri
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility, London, United Kingdom
- Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Bryan Williams
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility, London, United Kingdom
- Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Sonia Gandhi
- The Francis Crick Institute, London, United Kingdom
- University College London, London, United Kingdom
| | - Adam J. Kucharski
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Charles Swanton
- The Francis Crick Institute, London, United Kingdom
- University College London, London, United Kingdom
| | - David L. V. Bauer
- The Francis Crick Institute, London, United Kingdom
- Genotype-to-Phenotype UK National Virology Consortium (G2P-UK)
| | - Emma C. Wall
- The Francis Crick Institute, London, United Kingdom
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility, London, United Kingdom
| |
Collapse
|
40
|
Middleton C, Larremore DB. Modeling the Transmission Mitigation Impact of Testing for Infectious Diseases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.09.22.23295983. [PMID: 37808825 PMCID: PMC10557819 DOI: 10.1101/2023.09.22.23295983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
A fundamental question of any program focused on the testing and timely diagnosis of a communicable disease is its effectiveness in reducing transmission. Here, we introduce testing effectiveness (TE)-the fraction by which testing and post-diagnosis isolation reduce transmission at the population scale-and a model that incorporates test specifications and usage, within-host pathogen dynamics, and human behaviors to estimate TE. Using TE to guide recommendations, we show that today's rapid diagnostics should be used immediately upon symptom onset to control influenza A and respiratory syncytial virus (RSV), but delayed by up to 2d to control omicron-era SARS-CoV-2. Furthermore, while rapid tests are superior to RT-qPCR for control of founder-strain SARS-CoV-2, omicron-era changes in viral kinetics and rapid test sensitivity cause a reversal, with higher TE for RT-qPCR despite longer turnaround times. Finally, we illustrate the model's flexibility by quantifying tradeoffs in the use of post-diagnosis testing to shorten isolation times.
Collapse
Affiliation(s)
- Casey Middleton
- Department of Computer Science, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Daniel B Larremore
- Department of Computer Science, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
- Santa Fe Institute, Santa Fe, NM, USA
| |
Collapse
|
41
|
Owens K, Esmaeili-Wellman S, Schiffer JT. Heterogeneous SARS-CoV-2 kinetics due to variable timing and intensity of immune responses. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.08.20.23294350. [PMID: 37662228 PMCID: PMC10473815 DOI: 10.1101/2023.08.20.23294350] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The viral kinetics of documented SARS-CoV-2 infections exhibit a high degree of inter-individual variability. We identified six distinct viral shedding patterns, which differed according to peak viral load, duration, expansion rate and clearance rate, by clustering data from 768 infections in the National Basketball Association cohort. Omicron variant infections in previously vaccinated individuals generally led to lower cumulative shedding levels of SARS-CoV-2 than other scenarios. We then developed a mechanistic mathematical model that recapitulated 1510 observed viral trajectories, including viral rebound and cases of reinfection. Lower peak viral loads were explained by a more rapid and sustained transition of susceptible cells to a refractory state during infection, as well as an earlier and more potent late, cytolytic immune response. Our results suggest that viral elimination occurs more rapidly during omicron infection, following vaccination, and following re-infection due to enhanced innate and acquired immune responses. Because viral load has been linked with COVID-19 severity and transmission risk, our model provides a framework for understanding the wide range of observed SARS-CoV-2 infection outcomes.
Collapse
Affiliation(s)
- Katherine Owens
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division
| | | | - Joshua T Schiffer
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division
- University of Washington, Department of Medicine
| |
Collapse
|
42
|
Gubbins S. Quantifying the relationship between within-host dynamics and transmission for viral diseases of livestock. J R Soc Interface 2024; 21:20230445. [PMID: 38379412 PMCID: PMC10879856 DOI: 10.1098/rsif.2023.0445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/18/2024] [Indexed: 02/22/2024] Open
Abstract
Understanding the population dynamics of an infectious disease requires linking within-host dynamics and between-host transmission in a quantitative manner, but this is seldom done in practice. Here a simple phenomenological model for viral dynamics within a host is linked to between-host transmission by assuming that the probability of transmission is related to log viral titre. Data from transmission experiments for two viral diseases of livestock, foot-and-mouth disease virus in cattle and swine influenza virus in pigs, are used to parametrize the model and, importantly, test the underlying assumptions. The model allows the relationship between within-host parameters and transmission to be determined explicitly through their influence on the reproduction number and generation time. Furthermore, these critical within-host parameters (time and level of peak titre, viral growth and clearance rates) can be computed from more complex within-host models, raising the possibility of assessing the impact of within-host processes on between-host transmission in a more detailed quantitative manner.
Collapse
Affiliation(s)
- Simon Gubbins
- The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF, UK
| |
Collapse
|
43
|
Prelog M, Jeske SD, Asam C, Fuchs A, Wieser A, Gall C, Wytopil M, Mueller-Schmucker SM, Beileke S, Goekkaya M, Kling E, Geldmacher C, Rubio-Acero R, Plank M, Christa C, Willmann A, Vu M, Einhauser S, Weps M, Lampl BMJ, Almanzar G, Kousha K, Schwägerl V, Liebl B, Weber B, Drescher J, Scheidt J, Gefeller O, Messmann H, Protzer U, Liese J, Hoelscher M, Wagner R, Überla K, Steininger P. Clinical and immunological benefits of full primary COVID-19 vaccination in individuals with SARS-CoV-2 breakthrough infections: A prospective cohort study in non-hospitalized adults. J Clin Virol 2024; 170:105622. [PMID: 38091664 DOI: 10.1016/j.jcv.2023.105622] [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: 07/19/2023] [Revised: 11/23/2023] [Accepted: 11/26/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND SARS-CoV-2 variants of concern (VOC) may result in breakthrough infections (BTIs) in vaccinated individuals. The aim of this study was to investigate the effects of full primary (two-dose) COVID-19 vaccination with wild-type-based SARS-CoV-2 vaccines on symptoms and immunogenicity of SARS-CoV-2 VOC BTIs. METHODS In a longitudinal multicenter controlled cohort study in Bavaria, Germany, COVID-19 vaccinated and unvaccinated non-hospitalized individuals were prospectively enrolled within 14 days of a PCR-confirmed SARS-CoV-2 infection. Individuals were visited weekly up to 4 times, performing a structured record of medical data and viral load assessment. SARS-CoV-2-specific antibody response was characterized by anti-spike-(S)- and anti-nucleocapsid-(N)-antibody concentrations, anti-S-IgG avidity and neutralization capacity. RESULTS A total of 300 individuals (212 BTIs, 88 non-BTIs) were included with VOC Alpha or Delta SARS-CoV-2 infections. Full primary COVID-19 vaccination provided a significant effectiveness against five symptoms (relative risk reduction): fever (33 %), cough (21 %), dysgeusia (22 %), dizziness (52 %) and nausea/vomiting (48 %). Full primary vaccinated individuals showed significantly higher 50 % inhibitory concentration (IC50) values against the infecting VOC compared to unvaccinated individuals at week 1 (269 vs. 56, respectively), and weeks 5-7 (1,917 vs. 932, respectively) with significantly higher relative anti-S-IgG avidity (78% vs. 27 % at week 4, respectively). CONCLUSIONS Full primary COVID-19 vaccination reduced symptom frequencies in non-hospitalized individuals with BTIs and elicited a more rapid and longer lasting neutralization capacity against the infecting VOC compared to unvaccinated individuals. These results support the recommendation to offer at least full primary vaccination to all adults to reduce disease severity caused by immune escape-variants.
Collapse
Affiliation(s)
- Martina Prelog
- Pediatric Rheumatology / Special Immunology, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Samuel D Jeske
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Claudia Asam
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Andre Fuchs
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| | - Andreas Wieser
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Christine Gall
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Monika Wytopil
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sandra M Mueller-Schmucker
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stephanie Beileke
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mehmet Goekkaya
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Institute of Environmental Medicine Helmholtz Zentrum München, German Research Center for Environmental Health, Augsburg, Germany
| | - Elisabeth Kling
- Institute of Laboratory Medicine and Microbiology University Hospital Augsburg, Augsburg, Germany
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Raquel Rubio-Acero
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Michael Plank
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Catharina Christa
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Annika Willmann
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Martin Vu
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Sebastian Einhauser
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Manuela Weps
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Benedikt M J Lampl
- Regensburg Department of Public Health, Division of Infection Control and Prevention, Regensburg, Germany; Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Giovanni Almanzar
- Pediatric Rheumatology / Special Immunology, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Kimia Kousha
- Pediatric Rheumatology / Special Immunology, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Valeria Schwägerl
- Pediatric Infectious Diseases, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Bernhard Liebl
- Bavarian Health and Food Safety Authority (LGL), Oberschleißheim, Germany
| | - Beatrix Weber
- Institute for Information Systems, University of Applied Sciences Hof, Hof, Germany
| | | | - Jörg Scheidt
- Institute for Information Systems, University of Applied Sciences Hof, Hof, Germany
| | - Olaf Gefeller
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Helmut Messmann
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany; Institute of Virology, Helmholtz Munich, Munich, Germany, and German Center for Infection Research, Munich partner site
| | - Johannes Liese
- Pediatric Infectious Diseases, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Ralf Wagner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany; Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Steininger
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
44
|
Farjo M, Koelle K, Martin MA, Gibson LL, Walden KKO, Rendon G, Fields CJ, Alnaji FG, Gallagher N, Luo CH, Mostafa HH, Manabe YC, Pekosz A, Smith RL, McManus DD, Brooke CB. Within-host evolutionary dynamics and tissue compartmentalization during acute SARS-CoV-2 infection. J Virol 2024; 98:e0161823. [PMID: 38174928 PMCID: PMC10805032 DOI: 10.1128/jvi.01618-23] [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: 10/16/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024] Open
Abstract
The global evolution of SARS-CoV-2 depends in part upon the evolutionary dynamics within individual hosts with varying immune histories. To characterize the within-host evolution of acute SARS-CoV-2 infection, we sequenced saliva and nasal samples collected daily from vaccinated and unvaccinated individuals early during infection. We show that longitudinal sampling facilitates high-confidence genetic variant detection and reveals evolutionary dynamics missed by less-frequent sampling strategies. Within-host dynamics in both unvaccinated and vaccinated individuals appeared largely stochastic; however, in rare cases, minor genetic variants emerged to frequencies sufficient for forward transmission. Finally, we detected significant genetic compartmentalization of viral variants between saliva and nasal swab sample sites in many individuals. Altogether, these data provide a high-resolution profile of within-host SARS-CoV-2 evolutionary dynamics.IMPORTANCEWe detail the within-host evolutionary dynamics of SARS-CoV-2 during acute infection in 31 individuals using daily longitudinal sampling. We characterized patterns of mutational accumulation for unvaccinated and vaccinated individuals, and observed that temporal variant dynamics in both groups were largely stochastic. Comparison of paired nasal and saliva samples also revealed significant genetic compartmentalization between tissue environments in multiple individuals. Our results demonstrate how selection, genetic drift, and spatial compartmentalization all play important roles in shaping the within-host evolution of SARS-CoV-2 populations during acute infection.
Collapse
Affiliation(s)
- Mireille Farjo
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Katia Koelle
- Department of Biology, Emory University, Atlanta, Georgia, USA
| | - Michael A. Martin
- Department of Biology, Emory University, Atlanta, Georgia, USA
- Population Biology, Ecology, and Evolution Graduate Program, Emory University, Atlanta, Georgia, USA
| | - Laura L. Gibson
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kimberly K. O. Walden
- High-Performance Biological Computing at the Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Gloria Rendon
- High-Performance Biological Computing at the Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Christopher J. Fields
- High-Performance Biological Computing at the Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Fadi G. Alnaji
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Nicholas Gallagher
- Division of Medical Microbiology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chun Huai Luo
- Division of Medical Microbiology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Heba H. Mostafa
- Division of Medical Microbiology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yukari C. Manabe
- Division of Infectious Disease, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Rebecca L. Smith
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - David D. McManus
- Division of Cardiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Christopher B. Brooke
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
45
|
Feng Y, Fan Y, Luo X, Ge J. A Wells-Riley based COVID-19 infectious risk assessment model combining both short range and room scale effects. BUILDING SIMULATION 2024; 17:93-111. [DOI: 10.1007/s12273-023-1060-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/13/2023] [Accepted: 06/29/2023] [Indexed: 01/05/2025]
|
46
|
Lingas G, Planas D, Péré H, Porrot F, Guivel-Benhassine F, Staropoli I, Duffy D, Chapuis N, Gobeaux C, Veyer D, Delaugerre C, Le Goff J, Getten P, Hadjadj J, Bellino A, Parfait B, Treluyer JM, Schwartz O, Guedj J, Kernéis S, Terrier B. Neutralizing Antibody Levels as a Correlate of Protection Against SARS-CoV-2 Infection: A Modeling Analysis. Clin Pharmacol Ther 2024; 115:86-94. [PMID: 37795693 DOI: 10.1002/cpt.3069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023]
Abstract
Although anti-severe acute respiratory syndrome-coronavirus 2 antibody kinetics have been described in large populations of vaccinated individuals, we still poorly understand how they evolve during a natural infection and how this impacts viral clearance. For that purpose, we analyzed the kinetics of both viral load and neutralizing antibody levels in a prospective cohort of individuals during acute infection with alpha variant. Using a mathematical model, we show that the progressive increase in neutralizing antibodies leads to a shortening of the half-life of both infected cells and infectious viral particles. We estimated that the neutralizing activity reached 90% of its maximal level within 11 days after symptom onset and could reduce the half-life of both infected cells and circulating virus by a 6-fold factor, thus playing a key role to achieve rapid viral clearance. Using this model, we conducted a simulation study to predict in a more general context the protection conferred by pre-existing neutralization titers, due to either vaccination or prior infection. We predicted that a neutralizing activity, as measured by 50% effective dose > 103 , could reduce by 46% the risk of having viral load detectable by standard polymerase chain reaction assays and by 98% the risk of having viral load above the threshold of infectiousness. Our model shows that neutralizing activity could be used to define correlates of protection against infection and transmission.
Collapse
Affiliation(s)
| | - Delphine Planas
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
- Vaccine Research Institute, Créteil, France
| | - Hélène Péré
- Virology Unit, Microbiology Department, APHP, Hôpital Européen Georges-Pompidou, Paris, France
- Université Paris Cité, INSERM UMRS1138 Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Françoise Porrot
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
| | | | - Isabelle Staropoli
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Nicolas Chapuis
- Assistance Publique-Hôpitaux de Paris, Centre-Université Paris Cité, Service d'hématologie biologique, Hôpital Cochin, Paris, France
| | - Camille Gobeaux
- Department of Automated Biology, CHU de Cochin, AP-HP, Paris, France
| | - David Veyer
- Virology Unit, Microbiology Department, APHP, Hôpital Européen Georges-Pompidou, Paris, France
- Université Paris Cité, INSERM UMRS1138 Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Constance Delaugerre
- Virology Department, AP-HP, Hôpital Saint-Louis, Paris, France
- Université Paris Cité, Inserm U944, Biology of Emerging Viruses, Paris, France
| | - Jérôme Le Goff
- Virology Department, AP-HP, Hôpital Saint-Louis, Paris, France
- Université Paris Cité, Inserm U976, INSIGHT Team, Paris, France
| | | | - Jérôme Hadjadj
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hôpital Cochin, Paris, France
| | - Adèle Bellino
- URC-CIC Paris Centre Necker/Cochin, AP-HP, Hôpital Cochin, Paris, France
| | - Béatrice Parfait
- Fédération des Centres de Ressources Biologiques - Plateformes de Ressources Biologiques AP-HP.Centre-Université Paris Cité, Centre de Ressources Biologiques Cochin, Hôpital Cochin, Paris, France
| | - Jean-Marc Treluyer
- Unité de Recherche clinique, Hôpital Cochin, AP-HP.Centre - Université de Paris, Paris, France
| | - Olivier Schwartz
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
- Vaccine Research Institute, Créteil, France
| | | | - Solen Kernéis
- Université Paris Cité, IAME, INSERM, Paris, France
- Equipe de Prévention du Risque Infectieux (EPRI), AP-HP, Hôpital Bichat, Paris, France
| | - Benjamin Terrier
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hôpital Cochin, Paris, France
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
47
|
Reeve J. De-stabilizing innate immunity in COVID-19: effects of its own positive feedback and erratic viraemia on the alternative pathway of complement. ROYAL SOCIETY OPEN SCIENCE 2024; 11:221597. [PMID: 38234438 PMCID: PMC10791537 DOI: 10.1098/rsos.221597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 12/08/2023] [Indexed: 01/19/2024]
Abstract
Complement provides powerful, fast responses in the human circulation to SARS-CoV-2 (COVID-19 virus) infection of the lower respiratory tract. COVID-19 effects were investigated in a revised human in silico Mass Action model of complement's alternative pathway (AP) responses. Bursts of newly circulating virions increased the fission of Complement protein C3 into C3a and C3b via stimulation of the lectin pathway or inhibited complement factor H. Viral reproduction sub-models incorporated smoothly exponential or step-wise exponential growth. Starting complement protein concentrations were drawn randomly from published normal male or female ranges and each infection model run for 10 days. C3 and factor B (FB) syntheses driven by Lectin Pathway stimulation led to declining plasma C3 and increasing FB concentrations. The C3-convertase concentration, a driver of viral elimination, could match viral growth over three orders of magnitude but near-complete exhaustion of circulating C3 was more prevalent with step-wise than with 'smooth' increases in viral stimulation. C3 exhaustion could be prolonged. Type 2 Diabetes and hypertension led to greatly increased peak C3-convertase concentrations, as did short-term variability of COVID-19 viraemia, pulmonary capillary clotting and secondary acidosis. Positive feedback in the AP greatly extends its response range at the expense of stability.
Collapse
Affiliation(s)
- Jonathan Reeve
- Senior Research Fellow, Nuffield Department of Orthopaedics, Rheumatological and Musculoskeletal Sciences, University of Oxford Botnar Research Centre, Windmill Road, Oxford OX3 7LD, UK
| |
Collapse
|
48
|
Zhang L, Cao H, Medlin K, Pearson J, Aristotelous AC, Chen A, Wessler T, Forest MG. Computational Modeling Insights into Extreme Heterogeneity in COVID-19 Nasal Swab Data. Viruses 2023; 16:69. [PMID: 38257769 PMCID: PMC10820884 DOI: 10.3390/v16010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/20/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024] Open
Abstract
Throughout the COVID-19 pandemic, an unprecedented level of clinical nasal swab data from around the globe has been collected and shared. Positive tests have consistently revealed viral titers spanning six orders of magnitude! An open question is whether such extreme population heterogeneity is unique to SARS-CoV-2 or possibly generic to viral respiratory infections. To probe this question, we turn to the computational modeling of nasal tract infections. Employing a physiologically faithful, spatially resolved, stochastic model of respiratory tract infection, we explore the statistical distribution of human nasal infections in the immediate 48 h of infection. The spread, or heterogeneity, of the distribution derives from variations in factors within the model that are unique to the infected host, infectious variant, and timing of the test. Hypothetical factors include: (1) reported physiological differences between infected individuals (nasal mucus thickness and clearance velocity); (2) differences in the kinetics of infection, replication, and shedding of viral RNA copies arising from the unique interactions between the host and viral variant; and (3) differences in the time between initial cell infection and the clinical test. Since positive clinical tests are often pre-symptomatic and independent of prior infection or vaccination status, in the model we assume immune evasion throughout the immediate 48 h of infection. Model simulations generate the mean statistical outcomes of total shed viral load and infected cells throughout 48 h for each "virtual individual", which we define as each fixed set of model parameters (1) and (2) above. The "virtual population" and the statistical distribution of outcomes over the population are defined by collecting clinically and experimentally guided ranges for the full set of model parameters (1) and (2). This establishes a model-generated "virtual population database" of nasal viral titers throughout the initial 48 h of infection of every individual, which we then compare with clinical swab test data. Support for model efficacy comes from the sampling of infection dynamics over the virtual population database, which reproduces the six-order-of-magnitude clinical population heterogeneity. However, the goal of this study is to answer a deeper biological and clinical question. What is the impact on the dynamics of early nasal infection due to each individual physiological feature or virus-cell kinetic mechanism? To answer this question, global data analysis methods are applied to the virtual population database that sample across the entire database and de-correlate (i.e., isolate) the dynamic infection outcome sensitivities of each model parameter. These methods predict the dominant, indeed exponential, driver of population heterogeneity in dynamic infection outcomes is the latency time of infected cells (from the moment of infection until onset of viral RNA shedding). The shedding rate of the viral RNA of infected cells in the shedding phase is a strong, but not exponential, driver of infection. Furthermore, the unknown timing of the nasal swab test relative to the onset of infection is an equally dominant contributor to extreme population heterogeneity in clinical test data since infectious viral loads grow from undetectable levels to more than six orders of magnitude within 48 h.
Collapse
Affiliation(s)
- Leyi Zhang
- Department of Mathematics and Carolina Center for Interdisciplinary Applied Mathematics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Han Cao
- Department of Mathematics and Carolina Center for Interdisciplinary Applied Mathematics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Karen Medlin
- Department of Mathematics and Carolina Center for Interdisciplinary Applied Mathematics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jason Pearson
- Department of Mathematics and Carolina Center for Interdisciplinary Applied Mathematics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Simulations Plus, Inc., 6 Davis Dr., Durham, NC 27709, USA
| | | | - Alexander Chen
- Department of Mathematics, California State University, Dominguez Hills, CA 90747, USA
| | - Timothy Wessler
- Department of Applied Mathematics, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - M. Gregory Forest
- Department of Mathematics and Carolina Center for Interdisciplinary Applied Mathematics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Departments of Applied Physical Sciences and Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
49
|
Mohammadi A, Chiang S, Li F, Wei F, Lau CS, Aziz M, Ibarrondo FJ, Fulcher JA, Yang OO, Chia D, Kim Y, Wong DT. Direct Detection of 4-Dimensions of SARS-CoV-2: Infection (vRNA), Infectivity (Antigen), Binding Antibody, and Functional Neutralizing Antibody in Saliva. RESEARCH SQUARE 2023:rs.3.rs-3745787. [PMID: 38234820 PMCID: PMC10793499 DOI: 10.21203/rs.3.rs-3745787/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
We developed a 4-parameter clinical assay using Electric Field Induced Release and Measurement (EFIRM) technology to simultaneously assess SARS-CoV-2 RNA (vRNA), nucleocapsid antigen, host binding (BAb) and neutralizing antibody (NAb) levels from a drop of saliva with performance that equals or surpasses current EUA-approved tests. The vRNA and antigen assays achieved lower limit of detection (LOD) of 100 copies/reaction and 3.5 TCID₅₀/mL, respectively. The vRNA assay differentiated between acutely infected (n=10) and infection-naïve patients (n=33) with an AUC of 0.9818, sensitivity of 90%, and specificity of 100%. The antigen assay similarly differentiated these patient populations with an AUC of 1.000. The BAb assay detected BAbs with an LOD of 39 pg/mL and distinguished acutely infected (n=35), vaccinated with prior infection (n=13), and vaccinated infection-naïve patients (n=13) from control (n=81) with AUC of 0.9481, 1.000, and 0.9962, respectively. The NAb assay detected NAbs with an LOD of 31.6 Unit/mL and differentiated between COVID-19 recovered or vaccinated patients (n=31) and pre-pandemic controls (n=60) with an AUC 0.923, sensitivity of 87.10%, and specificity of 86.67%. Our multiparameter assay represents a significant technological advancement to simultaneously address SARS-CoV-2 infection and immunity, and it lays the foundation for tackling potential future pandemics.
Collapse
Affiliation(s)
- Aida Mohammadi
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Samantha Chiang
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Feng Li
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Fang Wei
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Mohammad Aziz
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Francisco J. Ibarrondo
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer A. Fulcher
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Otto O. Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - David Chia
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Yong Kim
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - David T.W. Wong
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
50
|
Nishiyama T, Miyamatsu Y, Park H, Nakamura N, Yokokawa Shibata R, Iwami S, Nagasaki Y. Modeling COVID-19 vaccine booster-elicited antibody response and impact of infection history. Vaccine 2023; 41:7655-7662. [PMID: 38008663 DOI: 10.1016/j.vaccine.2023.11.040] [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: 09/10/2023] [Revised: 10/28/2023] [Accepted: 11/18/2023] [Indexed: 11/28/2023]
Abstract
The 3-dose COVID-19 vaccine (booster vaccination) has been offered worldwide. As booster vaccinations continue, it is important to understand the antibody dynamics elicited by booster vaccination in order to evaluate and develop vaccination needs and strategies. Here, we investigated longitudinal data by monitoring IgG antibodies against the receptor binding domain (RBD) in health care workers. We extended our previously developed mathematical model to booster vaccines and successfully fitted antibody titers over time in the absence and presence of past SARS-CoV-2 infection. Quantitative analysis using our mathematical model indicated that anti-RBD IgG titers increase to a comparable extent after booster vaccination, regardless of the presence or absence of infection, but infection history extends the duration of antibody response by 1.28 times. Such a mathematical modeling approach can be used to inform future vaccination strategies on the basis of an individual's immune history. Our simple quantitative approach can be extended to any kind of vaccination and therefore can form a basis for policy decisions regarding the distribution of booster vaccines to strengthen immunity in future pandemics.
Collapse
Affiliation(s)
- Takara Nishiyama
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Yuichiro Miyamatsu
- Department of Neurosurgery, National Hospital Organization Kyushu Medical Center, Fukuoka 810-8563, Japan; Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-0054, Japan
| | - Hyeongki Park
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Naotoshi Nakamura
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Risa Yokokawa Shibata
- Department of Advanced Transdisciplinary Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Shingo Iwami
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan; Institute of Mathematics for Industry, Kyushu University, Fukuoka 819-0395, Japan; Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto 606-8501, Japan; NEXT-Ganken Program, Japanese Foundation for Cancer Research (JFCR), Tokyo 135-8550, Japan; Interdisciplinary Theoretical and Mathematical Sciences (iTHEMS), RIKEN, Wako 351-0198, Japan; Science Groove Inc., Fukuoka 810-0041, Japan.
| | - Yoji Nagasaki
- Department of Infectious Disease, Clinical Research Institute, National Hospital Organization Kyushu Medical Center,1-8-1 Jigyohama, Chuo-ku, Fukuoka 810-8563, Japan.
| |
Collapse
|