1
|
Li R, Zhao X, Jiang K, Tang J, Yang S, Hu J, Ma X. Performance of the Flash10 COVID-19 point-of-care molecular test. Sci Rep 2024; 14:25622. [PMID: 39465327 PMCID: PMC11514266 DOI: 10.1038/s41598-024-77837-1] [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: 07/29/2024] [Accepted: 10/25/2024] [Indexed: 10/29/2024] Open
Abstract
After the COVID-19 pandemic, fever clinics urgently require rapid nucleic acid tests to enhance their capacity for timely pathogen detection. This study evaluated the analytical performance and clinical utility of the Flash10 SARS-CoV-2 point-of-care test (Flash10 POCT) for detecting SARS-CoV-2 in patients with fever in the adult fever clinic in Beijing Tsinghua Changgung Hospital from August 1 to August 30, 2023. The analytical performance and clinical utility of the Flash10 POCT for detecting SARS-CoV-2 were assessed in 125 patients with fever syndrome in the adult fever clinic. The Flash10 POCT demonstrated an analytical precision of 3.1% for the Ct values of the ORF1ab gene and 2.9% for the Ct values of the N gene in SARS-CoV-2 nucleic acid testing. Furthermore, the Flash10 POCT demonstrated a lower limit of detection (LoD) of 100 copies/mL, with no detected aerosol contamination leakage. Of the 125 patients (median age 61.9 years, 52% male and 48% female), both the Flash10 POCT and RT-PCR tests yielded positive results for 100 patients and negative results for 25 patients (Fisher's exact test, p < 0.0001). The median turn-around-time for the Flash10 POCT was significantly shorter, at 1.05 h, compared to 16.15 h required for RT-PCR tests (Wilcoxon signed rank test, p < 0.0001). The Flash10 POCT showed high analytical performance, achieving a 100% detection rate for SARS-CoV-2 compared to RT-PCR tests, while also exhibiting a significantly shorter turn-around-time. Implementing the Flash10 POCT had the potential to expedite the care of adults presenting with fever.
Collapse
Affiliation(s)
- Runqing Li
- Laboratory Medicine Department of Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, No. 168 Litang Road, Changping District, Beijing, 102218, China.
- Laboratory Medicine Department of Tiantongyuan North Community Healthcare Center, Beijing, China.
| | - Xiuying Zhao
- Laboratory Medicine Department of Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, No. 168 Litang Road, Changping District, Beijing, 102218, China
| | - Kai Jiang
- Laboratory Medicine Department of Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, No. 168 Litang Road, Changping District, Beijing, 102218, China
| | - Jie Tang
- Laboratory Medicine Department of Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, No. 168 Litang Road, Changping District, Beijing, 102218, China
| | - Song Yang
- Laboratory Medicine Department of Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, No. 168 Litang Road, Changping District, Beijing, 102218, China
| | - Jing Hu
- Laboratory Medicine Department of Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, No. 168 Litang Road, Changping District, Beijing, 102218, China
| | - Xuzhu Ma
- Infectious Disease Department of Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
2
|
Seaman WT, Keener O, Nanayakkara D, Mollan KR, Premkumar L, Cuadra EC, Jones CD, Pettifor A, Bowman NM, Wang F, Webster-Cyriaque J. Oral SARS-CoV-2 host responses predict the early COVID-19 disease course. Sci Rep 2024; 14:21788. [PMID: 39294156 PMCID: PMC11411107 DOI: 10.1038/s41598-024-67504-w] [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: 07/09/2023] [Accepted: 07/11/2024] [Indexed: 09/20/2024] Open
Abstract
Oral fluids provide ready detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses. This study sought to evaluate relationships between oral virus, oral and systemic anti-SARS-CoV-2-specific antibodies, and symptoms. Oral fluids (saliva/throat wash (saliva/TW)) and serum were collected from asymptomatic and symptomatic, nasopharyngeal (NP) SARS-CoV-2 RT-qPCR+ human participants (n = 45). SARS-CoV-2 RT-qPCR and N-antigen detection by immunoblot and lateral flow assay (LFA) were performed. RT-qPCR for subgenomic RNA (sgRNA) was sequence confirmed. SARS-CoV-2-anti-S protein RBD LFA and ELISA assessed IgM and IgG responses. Structural analysis identified host salivary molecules analogous to SARS-CoV-2-N-antigen. At time of enrollment (baseline, BL), LFA-detected N-antigen in 86% of TW and was immunoblot-confirmed. However, only 3/17 were saliva/TW qPCR+ . Sixty percent of saliva and 83% of TW demonstrated persistent N-antigen at 4 weeks. N-antigen LFA signal in three anti-spike sero-negative participants suggested potential cross-detection of 4 structurally analogous salivary RNA binding proteins (alignment 19-29aa, RMSD 1-1.5 Angstroms). At enrollment, symptomatic participants demonstrated replication-associated sgRNA junctions, were IgG+ (94%/100% in saliva/TW), and IgM+ (63%/54%). At 4 weeks, SARS-CoV-2 IgG (100%/83%) and IgM (80%/67%) persisted. Oral and serum IgG correlated 100% with NP+ PCR status. Cough and fatigue severity (p = 0.010 and 0.018 respectively), and presence of weakness, nausea, and composite upper respiratory symptoms (p = 0.037, 0.005, and 0.017, respectively) were negatively associated with saliva IgM but not TW or serum IgM. Throat wash IgM levels were higher in women compared to men, although the association did not reach statistical significance (median: 290 (female) versus 0.697, p = 0.056). Important to transmission and disease course, oral viral replication and persistence showed clear relationships with select symptoms and early oral IgM responses during early infection. N-antigen cross-reactivity may reflect mimicry of structurally analogous host proteins.
Collapse
Affiliation(s)
- William T Seaman
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Olive Keener
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- North Carolina School of Math and Science, Durham, NC, USA
| | - Dinelka Nanayakkara
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katie R Mollan
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Edwing Centeno Cuadra
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Corbin D Jones
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Audrey Pettifor
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Natalie M Bowman
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
| | | | - Jennifer Webster-Cyriaque
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA.
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
3
|
Grimm M, Ziegler L, Seglias A, Mademilov M, Magdieva K, Mirzalieva G, Taalaibekova A, Suter S, Schneider SR, Zoller F, Bissig V, Reinhard L, Bauer M, Müller J, Ulrich TL, Carta AF, Bader PR, Bitos K, Reiser AE, Champigneulle B, Ashyralieva D, Scheiwiller PM, Ulrich S, Sooronbaev TM, Furian M, Bloch KE. SARS-CoV-2 Transmission during High-Altitude Field Studies. High Alt Med Biol 2024; 25:197-204. [PMID: 38634740 DOI: 10.1089/ham.2023.0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024] Open
Abstract
Grimm, Mirjam, Lucie Ziegler, Annina Seglias, Maamed Mademilov, Kamila Magdieva, Gulzada Mirzalieva, Aijan Taalaibekova, Simone Suter, Simon R. Schneider, Fiona Zoller, Vera Bissig, Lukas Reinhard, Meret Bauer, Julian Müller, Tanja L. Ulrich, Arcangelo F. Carta, Patrick R. Bader, Konstantinos Bitos, Aurelia E. Reiser, Benoit Champigneulle, Damira Ashyralieva, Philipp M. Scheiwiller, Silvia Ulrich, Talant M. Sooronbaev, Michael Furian, and Konrad E. Bloch. SARS-CoV-2 Transmission during High-Altitude Field Studies. High Alt Med Biol. 25:197-204, 2024. Background: Throughout the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) pandemic, virus transmission during clinical research was of concern. Therefore, during high-altitude field studies performed in 2021, we took specific COVID-19 precautions and investigated the occurrence of SARS-CoV-2 infection. Methods: From May to September 2021, we performed studies in patients with chronic obstructive pulmonary disease (COPD) and in healthy school-age children in Kyrgyzstan in high-altitude facilities at 3,100 m and 3,250 m and at 760 m. The various implemented COVID-19 safety measures included systematic SARS-CoV-2 rapid antigen testing (RAT). Main outcomes were SARS-CoV-2-RAT-positive rate among participants and staff at initial presentation (prevalence) and SARS-CoV-2-RAT-positive conversion during and within 10 days after studies (incidence). Results: Among 338 participants and staff, SARS-CoV-2-RAT-positive prevalence was 15 (4.4%). During mean ± SD duration of individual study participation of 3.1 ± 1.0 day and within 10 days, RAT-positive conversion occurred in 1/237(0.4%) participants. Among staff working in studies for 31.5 ± 29.3 days, SARS-CoV-2-RAT-positive conversion was 11/101(10.9%). In all 338 individuals involved in the studies over the course of 15.6 weeks, the median SARS-CoV-2-RAT-positive incidence was 0.00%/week (quartiles 0.00; 0.64). Over the same period, the median background incidence among the total Kyrgyz population of 6,636 million was 0.06%/week (0.03; 0.11), p = 0.013 (Wilcoxon rank sum test). Conclusions: Taking precautions by implementing specific safety measures, SARS-CoV-2 transmission during clinical studies was very rare, and the SARS-CoV-2 incidence among participants and staff was lower than that in the general population during the same period. The results are reassuring and may help in decision-making on the conduct of clinical research in similar settings.
Collapse
Affiliation(s)
- Mirjam Grimm
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Lucie Ziegler
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Annina Seglias
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Maamed Mademilov
- Department of Respiratory Medicine, National Center for Cardiology and Internal Medicine, Bishkek, Kyrgyz Republic
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Kamila Magdieva
- Department of Respiratory Medicine, National Center for Cardiology and Internal Medicine, Bishkek, Kyrgyz Republic
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Gulzada Mirzalieva
- Department of Respiratory Medicine, National Center for Cardiology and Internal Medicine, Bishkek, Kyrgyz Republic
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Aijan Taalaibekova
- Department of Respiratory Medicine, National Center for Cardiology and Internal Medicine, Bishkek, Kyrgyz Republic
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Simone Suter
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Simon R Schneider
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Fiona Zoller
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Vera Bissig
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Lukas Reinhard
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Meret Bauer
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Julian Müller
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Tanja L Ulrich
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Arcangelo F Carta
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Patrick R Bader
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Konstantinos Bitos
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Aurelia E Reiser
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | | | - Damira Ashyralieva
- National Institute of Public Health, Ministry of Health, Bishkek, Kyrgyz Republic
| | - Philipp M Scheiwiller
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Silvia Ulrich
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Talant M Sooronbaev
- Department of Respiratory Medicine, National Center for Cardiology and Internal Medicine, Bishkek, Kyrgyz Republic
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Michael Furian
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| | - Konrad E Bloch
- Department of Respiratory Medicine, University Hospital of Zurich, Zurich, Switzerland
- Swiss-Kyrgyz High Altitude Medicine and Research Initiative, Zurich, Switzerland
| |
Collapse
|
4
|
Liang J, Wang T, Wang Q, Wang X, Fan X, Hu T, Leng X, Shi K, Li J, Gong Q, Du R. Prevalence of canine distemper in minks, foxes and raccoon dogs from 1983 to 2023 in Asia, North America, South America and Europe. Front Vet Sci 2024; 11:1394631. [PMID: 39193367 PMCID: PMC11348944 DOI: 10.3389/fvets.2024.1394631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Canine distemper (CD) is a virulent disease caused by the canine distemper virus (CDV) in canines and mustelidaes with high mortality. The incidence of CDV is worldwide distribution and it has caused huge economic losses to multiple industries around the world. There are many studies investigating the prevalence of CD infection, but no comprehensive analysis of CDV infection in minks, foxes and raccoon dogs worldwide has therefore been carried out. The aim of this meta is to provide a comprehensive assessment of the prevalence of CDV infection in minks, foxes and raccoon dogs dogs through a meta-analysis of articles published from around the world. Data from 8,582 small carnivores in 12 countries were used to calculate the combined prevalence of CD. A total of 22.6% (1,937/8,582) of minks, foxes and raccoon dogs tested positive for CD. The prevalence was higher in Asia (13.8, 95% CI: 22.2-45.6), especially in South Korea (65.8, 95% CI: 83.3-95.8). Our study found that the incidence of CD was also associated with geographic climate, population size, health status, and breeding patterns. CD is more commonly transmitted in minks, foxes and raccoon dogs. However, the concentrated breeding as an economic animal has led to an increase in the prevalence rate. The difference analysis study recommended that countries develop appropriate preventive and control measures based on the prevalence in the minks, foxes, and raccoon dogs industries, and that reducing stocking density is important to reduce the incidence of CDV. In addition, CDV is more common in winter, so vaccination in winter should be strengthened and expanded to reduce the incidence of CD in minks, foxes and raccoon dogs.
Collapse
Affiliation(s)
- Jian Liang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Tingting Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Qi Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Xiaolin Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Xinying Fan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Tingting Hu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Xue Leng
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Kun Shi
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Jianming Li
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Qinglong Gong
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Rui Du
- Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, Jilin, China
| |
Collapse
|
5
|
De Rop L, Bos DA, Stegeman I, Holtman G, Ochodo EA, Spijker R, Otieno JA, Alkhlaileh F, Deeks JJ, Dinnes J, Van den Bruel A, McInnes MD, Leeflang MM, Verbakel JY. Accuracy of routine laboratory tests to predict mortality and deterioration to severe or critical COVID-19 in people with SARS-CoV-2. Cochrane Database Syst Rev 2024; 8:CD015050. [PMID: 39105481 PMCID: PMC11301994 DOI: 10.1002/14651858.cd015050.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
BACKGROUND Identifying patients with COVID-19 disease who will deteriorate can be useful to assess whether they should receive intensive care, or whether they can be treated in a less intensive way or through outpatient care. In clinical care, routine laboratory markers, such as C-reactive protein, are used to assess a person's health status. OBJECTIVES To assess the accuracy of routine blood-based laboratory tests to predict mortality and deterioration to severe or critical (from mild or moderate) COVID-19 in people with SARS-CoV-2. SEARCH METHODS On 25 August 2022, we searched the Cochrane COVID-19 Study Register, encompassing searches of various databases such as MEDLINE via PubMed, CENTRAL, Embase, medRxiv, and ClinicalTrials.gov. We did not apply any language restrictions. SELECTION CRITERIA We included studies of all designs that produced estimates of prognostic accuracy in participants who presented to outpatient services, or were admitted to general hospital wards with confirmed SARS-CoV-2 infection, and studies that were based on serum banks of samples from people. All routine blood-based laboratory tests performed during the first encounter were included. We included any reference standard used to define deterioration to severe or critical disease that was provided by the authors. DATA COLLECTION AND ANALYSIS Two review authors independently extracted data from each included study, and independently assessed the methodological quality using the Quality Assessment of Prognostic Accuracy Studies tool. As studies reported different thresholds for the same test, we used the Hierarchical Summary Receiver Operator Curve model for meta-analyses to estimate summary curves in SAS 9.4. We estimated the sensitivity at points on the SROC curves that corresponded to the median and interquartile range boundaries of specificities in the included studies. Direct and indirect comparisons were exclusively conducted for biomarkers with an estimated sensitivity and 95% CI of ≥ 50% at a specificity of ≥ 50%. The relative diagnostic odds ratio was calculated as a summary of the relative accuracy of these biomarkers. MAIN RESULTS We identified a total of 64 studies, including 71,170 participants, of which 8169 participants died, and 4031 participants deteriorated to severe/critical condition. The studies assessed 53 different laboratory tests. For some tests, both increases and decreases relative to the normal range were included. There was important heterogeneity between tests and their cut-off values. None of the included studies had a low risk of bias or low concern for applicability for all domains. None of the tests included in this review demonstrated high sensitivity or specificity, or both. The five tests with summary sensitivity and specificity above 50% were: C-reactive protein increase, neutrophil-to-lymphocyte ratio increase, lymphocyte count decrease, d-dimer increase, and lactate dehydrogenase increase. Inflammation For mortality, summary sensitivity of a C-reactive protein increase was 76% (95% CI 73% to 79%) at median specificity, 59% (low-certainty evidence). For deterioration, summary sensitivity was 78% (95% CI 67% to 86%) at median specificity, 72% (very low-certainty evidence). For the combined outcome of mortality or deterioration, or both, summary sensitivity was 70% (95% CI 49% to 85%) at median specificity, 60% (very low-certainty evidence). For mortality, summary sensitivity of an increase in neutrophil-to-lymphocyte ratio was 69% (95% CI 66% to 72%) at median specificity, 63% (very low-certainty evidence). For deterioration, summary sensitivity was 75% (95% CI 59% to 87%) at median specificity, 71% (very low-certainty evidence). For mortality, summary sensitivity of a decrease in lymphocyte count was 67% (95% CI 56% to 77%) at median specificity, 61% (very low-certainty evidence). For deterioration, summary sensitivity of a decrease in lymphocyte count was 69% (95% CI 60% to 76%) at median specificity, 67% (very low-certainty evidence). For the combined outcome, summary sensitivity was 83% (95% CI 67% to 92%) at median specificity, 29% (very low-certainty evidence). For mortality, summary sensitivity of a lactate dehydrogenase increase was 82% (95% CI 66% to 91%) at median specificity, 60% (very low-certainty evidence). For deterioration, summary sensitivity of a lactate dehydrogenase increase was 79% (95% CI 76% to 82%) at median specificity, 66% (low-certainty evidence). For the combined outcome, summary sensitivity was 69% (95% CI 51% to 82%) at median specificity, 62% (very low-certainty evidence). Hypercoagulability For mortality, summary sensitivity of a d-dimer increase was 70% (95% CI 64% to 76%) at median specificity of 56% (very low-certainty evidence). For deterioration, summary sensitivity was 65% (95% CI 56% to 74%) at median specificity of 63% (very low-certainty evidence). For the combined outcome, summary sensitivity was 65% (95% CI 52% to 76%) at median specificity of 54% (very low-certainty evidence). To predict mortality, neutrophil-to-lymphocyte ratio increase had higher accuracy compared to d-dimer increase (RDOR (diagnostic Odds Ratio) 2.05, 95% CI 1.30 to 3.24), C-reactive protein increase (RDOR 2.64, 95% CI 2.09 to 3.33), and lymphocyte count decrease (RDOR 2.63, 95% CI 1.55 to 4.46). D-dimer increase had higher accuracy compared to lymphocyte count decrease (RDOR 1.49, 95% CI 1.23 to 1.80), C-reactive protein increase (RDOR 1.31, 95% CI 1.03 to 1.65), and lactate dehydrogenase increase (RDOR 1.42, 95% CI 1.05 to 1.90). Additionally, lactate dehydrogenase increase had higher accuracy compared to lymphocyte count decrease (RDOR 1.30, 95% CI 1.13 to 1.49). To predict deterioration to severe disease, C-reactive protein increase had higher accuracy compared to d-dimer increase (RDOR 1.76, 95% CI 1.25 to 2.50). The neutrophil-to-lymphocyte ratio increase had higher accuracy compared to d-dimer increase (RDOR 2.77, 95% CI 1.58 to 4.84). Lastly, lymphocyte count decrease had higher accuracy compared to d-dimer increase (RDOR 2.10, 95% CI 1.44 to 3.07) and lactate dehydrogenase increase (RDOR 2.22, 95% CI 1.52 to 3.26). AUTHORS' CONCLUSIONS Laboratory tests, associated with hypercoagulability and hyperinflammatory response, were better at predicting severe disease and mortality in patients with SARS-CoV-2 compared to other laboratory tests. However, to safely rule out severe disease, tests should have high sensitivity (> 90%), and none of the identified laboratory tests met this criterion. In clinical practice, a more comprehensive assessment of a patient's health status is usually required by, for example, incorporating these laboratory tests into clinical prediction rules together with clinical symptoms, radiological findings, and patient's characteristics.
Collapse
Affiliation(s)
- Liselore De Rop
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - David Ag Bos
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Inge Stegeman
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Utrecht, Utrecht, Netherlands
- Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Gea Holtman
- Department of Primary- and Long-term Care, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Eleanor A Ochodo
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
- Centre for Evidence-based Health Care, Department of Global Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - René Spijker
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Medical Library, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health, Amsterdam, Netherlands
| | - Jenifer A Otieno
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Fade Alkhlaileh
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Jonathan J Deeks
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Jacqueline Dinnes
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Ann Van den Bruel
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Matthew Df McInnes
- Department of Radiology, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Canada
| | - Mariska Mg Leeflang
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Jan Y Verbakel
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
Chen M, Yang Z, Gong H, Wu H, Liu L, Jiang JS, Gao JH, Tang CW, Huang ZY. Long-term intermittent oral administration of selective COX-2 inhibitor improved the clinical outcomes of COVID-19 in patients with cirrhosis. J Dig Dis 2024; 25:517-524. [PMID: 39350571 DOI: 10.1111/1751-2980.13313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVES Patients with cirrhosis are more susceptible to coronavirus disease 2019 (COVID-19) due to immune dysfunction. In this retrospective study we aimed to investigate whether suppression of mild systemic inflammation with selective cyclooxygenase-2 inhibitor (COX-2-I) during chronic care of cirrhotic patients would reduce the occurrence of acute decompensated events and improve patient prognosis of COVID-19. METHODS Medical records of cirrhotic patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection were sequentially reviewed. The patients were divided into the COX-2-I and control groups depending on whether they took oral selective COX-2-I for over 3 months or not. The primary outcomes included the occurrence of severe/critical COVID-19, acute decompensated events, and acute-on-chronic liver failure (ACLF). RESULTS After propensity score matching analysis, there were 314 cases in the control group and 118 cases in the COX-2-I group. Compared with the control group, the risk of severe/critical COVID-19 in the COX-2-I group was significantly decreased by 83.1% (p = 0.004). Acute decompensated events and ACLF occurred in 23 (7.32%) and nine (2.87%) cases in the control group, but none in the COX-2-I group (p = 0.003 and 0.122). The rate of hospitalization in the COX-2-I group was significantly lower than that of the control group (3.39% vs 13.06%, p = 0.003). No patient in the COX-2-I group required intensive care unit admission. CONCLUSIONS Long-term intermittent oral administration of selective COX-2-I in cirrhotic patients significantly reduces the occurrence of severe/critical COVID-19, acute decompensated events, and ACLF. It may also be used for systemic inflammation caused by other pathogens.
Collapse
Affiliation(s)
- Ming Chen
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhu Yang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hui Gong
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hao Wu
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ling Liu
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jing Sun Jiang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jin Hang Gao
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Cheng Wei Tang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhi Yin Huang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
7
|
Fomenko A, Dähne T, Weibel S, Panning M, Grummich K, Schlesinger S, Rücker G, Hengel H. Test accuracy of rapid diagnostic tests and reverse-transcription polymerase chain reaction against virus isolation in cell culture for assessing SARS-CoV-2 infectivity: Systematic review and meta-analysis. Rev Med Virol 2024; 34:e2569. [PMID: 38986606 DOI: 10.1002/rmv.2569] [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: 01/25/2024] [Revised: 04/25/2024] [Accepted: 06/23/2024] [Indexed: 07/12/2024]
Abstract
We aimed to assess the performance of Ag-RDT and RT-qPCR with regard to detecting infectious SARS-CoV-2 in cell cultures, as their diagnostic test accuracy (DTA) compared to virus isolation remains largely unknown. We searched three databases up to 15 December 2021 for DTA studies. The bivariate model was used to synthesise the estimates. Risk of bias was assessed using QUADAS-2/C. Twenty studies (2605 respiratory samples) using cell culture and at least one molecular test were identified. All studies were at high or unclear risk of bias in at least one domain. Three comparative DTA studies reported results on Ag-RDT and RT-qPCR against cell culture. Two studies evaluated RT-qPCR against cell culture only. Fifteen studies evaluated Ag-RDT against cell culture as reference standard in RT-qPCR-positive samples. For Ag-RDT, summary sensitivity was 93% (95% CI 78; 98%) and specificity 87% (95% CI 70; 95%). For RT-qPCR, summary sensitivity (continuity-corrected) was 98% (95% CI 95; 99%) and specificity 45% (95% CI 28; 63%). In studies relying on RT-qPCR-positive subsamples (n = 15), the summary sensitivity of Ag-RDT was 93% (95% CI 92; 93%) and specificity 63% (95% CI 63; 63%). Ag-RDT show moderately high sensitivity, detecting most but not all samples demonstrated to be infectious based on virus isolation. Although RT-qPCR exhibits high sensitivity across studies, its low specificity to indicate infectivity raises the question of its general superiority in all clinical settings. Study findings should be interpreted with caution due to the risk of bias, heterogeneity and the imperfect reference standard for infectivity.
Collapse
Affiliation(s)
- Alexey Fomenko
- Institute of Virology, Medical Center, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department Clinical Medicine, Institute of General Practice and Health Services Research, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Theo Dähne
- Institute of Virology, Medical Center, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Stephanie Weibel
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Marcus Panning
- Institute of Virology, Medical Center, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Kathrin Grummich
- Institute for Evidence in Medicine, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
| | - Sabrina Schlesinger
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DDZ), Partner Düsseldorf, Munich-Neuherberg, Germany
| | - Gerta Rücker
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
8
|
Madewell Z, Major C, Graff N, Adams C, Rodriguez D, Morales T, Medina Lopes N, Tosado R, Sánchez‐González L, Perez‐Padilla J, Volkman H, Bertrán‐Pasarell J, Sainz de la Peña D, Munoz‐Jordan J, Santiago G, Lorenzi O, Rivera‐Amill V, Rolfes M, Paz‐Bailey G, Adams L, Wong J. Diagnostic Accuracy of the Abbott BinaxNOW COVID-19 Antigen Card Test, Puerto Rico. Influenza Other Respir Viruses 2024; 18:e13305. [PMID: 39053895 PMCID: PMC11300111 DOI: 10.1111/irv.13305] [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: 12/15/2023] [Revised: 03/21/2024] [Accepted: 04/17/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic underscored the need for rapid and accurate diagnostic tools. In August 2020, the Abbott BinaxNOW COVID-19 Antigen Card test became available as a timely and affordable alternative for SARS-CoV-2 molecular testing, but its performance may vary due to factors including timing and symptomatology. This study evaluates BinaxNOW diagnostic performance in diverse epidemiological contexts. METHODS Using RT-PCR as reference, we assessed performance of the BinaxNOW COVID-19 test for SARS-CoV-2 detection in anterior nasal swabs from participants of two studies in Puerto Rico from December 2020 to May 2023. Test performance was assessed by days post symptom onset, collection strategy, vaccination status, symptomatology, repeated testing, and RT-PCR cycle threshold (Ct) values. RESULTS BinaxNOW demonstrated an overall sensitivity of 84.1% and specificity of 98.8%. Sensitivity peaked within 1-6 days after symptom onset (93.2%) and was higher for symptomatic (86.3%) than asymptomatic (67.3%) participants. Sensitivity declined over the course of infection, dropping from 96.3% in the initial test to 48.4% in testing performed 7-14 days later. BinaxNOW showed 99.5% sensitivity in participants with low Ct values (≤ 25) but lower sensitivity (18.2%) for participants with higher Cts (36-40). CONCLUSIONS BinaxNOW demonstrated high sensitivity and specificity, particularly in early-stage infections and symptomatic participants. In situations where test sensitivity is crucial for clinical decision-making, nucleic acid amplification tests are preferred. These findings highlight the importance of considering clinical and epidemiological context when interpreting test results and emphasize the need for ongoing research to adapt testing strategies to emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Zachary J. Madewell
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | - Chelsea G. Major
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | - Nathan Graff
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | - Cameron Adams
- Department of Microbiology and ImmunologyUniversity of North Carolina School of MedicineChapel HillNorth CarolinaUSA
| | - Dania M. Rodriguez
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | - Tatiana Morales
- Ponce Research InstitutePonce Health Sciences UniversityPoncePuerto Rico
| | - Nicole A. Medina Lopes
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | - Rafael Tosado
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | | | - Janice Perez‐Padilla
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | - Hannah R. Volkman
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | | | | | - Jorge Munoz‐Jordan
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | - Gilberto A. Santiago
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | - Olga Lorenzi
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | | | - Melissa A. Rolfes
- Influenza DivisionCenters for Disease Control and PreventionAtlantaGAUSA
| | - Gabriela Paz‐Bailey
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | - Laura E. Adams
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| | - Joshua M. Wong
- Division of Vector‐Borne DiseasesCenters for Disease Control and PreventionSan JuanPuerto Rico
| |
Collapse
|
9
|
Lane D, Allsopp R, Holmes CW, Slingsby OC, Jukes-Jones R, Bird P, Anderson NL, Razavi M, Yip R, Pearson TW, Pope M, Khunti K, Doykov I, Hällqvist J, Mills K, Skipp P, Carling R, Ng L, Shaw J, Gupta P, Jones DJL. A high throughput immuno-affinity mass spectrometry method for detection and quantitation of SARS-CoV-2 nucleoprotein in human saliva and its comparison with RT-PCR, RT-LAMP, and lateral flow rapid antigen test. Clin Chem Lab Med 2024; 62:1206-1216. [PMID: 38253336 DOI: 10.1515/cclm-2023-0243] [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: 03/06/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024]
Abstract
OBJECTIVES Many reverse transcription polymerase chain reaction (RT-PCR) methods exist that can detect SARS-CoV-2 RNA in different matrices. RT-PCR is highly sensitive, although viral RNA may be detected long after active infection has taken place. SARS-CoV-2 proteins have shorter detection windows hence their detection might be more meaningful. Given salivary droplets represent a main source of transmission, we explored the detection of viral RNA and protein using four different detection platforms including SISCAPA peptide immunoaffinity liquid chromatography-mass spectrometry (SISCAPA-LC-MS) using polyclonal capture antibodies. METHODS The SISCAPA-LC MS method was compared to RT-PCR, RT-loop-mediated isothermal amplification (RT-LAMP), and a lateral flow rapid antigen test (RAT) for the detection of virus material in the drool saliva of 102 patients hospitalised after infection with SARS-CoV-2. Cycle thresholds (Ct) of RT-PCR (E gene) were compared to RT-LAMP time-to-positive (TTP) (NE and Orf1a genes), RAT optical densitometry measurements (test line/control line ratio) and to SISCAPA-LC-MS for measurements of viral protein. RESULTS SISCAPA-LC-MS showed low sensitivity (37.7 %) but high specificity (89.8 %). RAT showed lower sensitivity (24.5 %) and high specificity (100 %). RT-LAMP had high sensitivity (83.0 %) and specificity (100.0 %). At high initial viral RNA loads (<20 Ct), results obtained using SISCAPA-LC-MS correlated with RT-PCR (R2 0.57, p-value 0.002). CONCLUSIONS Detection of SARS-CoV-2 nucleoprotein in saliva was less frequent than the detection of viral RNA. The SISCAPA-LC-MS method allowed processing of multiple samples in <150 min and was scalable, enabling high throughput.
Collapse
Affiliation(s)
- Dan Lane
- The Department of Chemical Pathology and Metabolic Diseases, Leicester Royal Infirmary, University Hospitals of Leicester, Leicester, UK
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Rebecca Allsopp
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Christopher W Holmes
- Clinical Microbiology, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, UK
| | | | - Rebekah Jukes-Jones
- The Department of Chemical Pathology and Metabolic Diseases, Leicester Royal Infirmary, University Hospitals of Leicester, Leicester, UK
| | - Paul Bird
- Clinical Microbiology, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, UK
| | | | | | - Richard Yip
- SISCAPA Assay Technologies, Inc., Washington, DC, USA
| | | | - Matt Pope
- SISCAPA Assay Technologies, Inc., Washington, DC, USA
| | - Kamlesh Khunti
- Leicester Diabetes Centre, Leicester General Hospital, University of Leicester, Leicester, UK
| | - Ivan Doykov
- Genetics & Genomic Medicine Department, Translational Mass Spectrometry Research Group, UCL Institute of Child Health, London, UK
- Great Ormond Street Biomedical Research Centre, UCL Institute of Child Health, London, UK
| | - Jenny Hällqvist
- Genetics & Genomic Medicine Department, Translational Mass Spectrometry Research Group, UCL Institute of Child Health, London, UK
- Great Ormond Street Biomedical Research Centre, UCL Institute of Child Health, London, UK
| | - Kevin Mills
- Genetics & Genomic Medicine Department, Translational Mass Spectrometry Research Group, UCL Institute of Child Health, London, UK
- Great Ormond Street Biomedical Research Centre, UCL Institute of Child Health, London, UK
| | - Paul Skipp
- Centre for Proteomic Research, University of Southampton, Southampton, UK
| | - Rachel Carling
- Biochemical Sciences, Synnovis, Guys & St Thomas' NHSFT, London, UK
- GKT School Medical Education, Kings College London, London, UK
| | - Leong Ng
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- van Geest MS-OMICS Facility, University of Leicester, Leicester, UK
| | - Jacqui Shaw
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Pankaj Gupta
- The Department of Chemical Pathology and Metabolic Diseases, Leicester Royal Infirmary, University Hospitals of Leicester, Leicester, UK
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Donald J L Jones
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
- van Geest MS-OMICS Facility, University of Leicester, Leicester, UK
| |
Collapse
|
10
|
McCracken GR, Patriquin G, Hatchette TF, Davidson RJ, Goodall B, Barrett L, MacDonald J, Heinstein C, Pettipas J, Ross J, LeBlanc JJ. Taqman PACMAN: a simple molecular approach for positive rapid antigen test confirmation during periods of low prevalence. Microbiol Spectr 2024; 12:e0407323. [PMID: 38567975 PMCID: PMC11064490 DOI: 10.1128/spectrum.04073-23] [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: 12/11/2023] [Accepted: 03/13/2024] [Indexed: 05/03/2024] Open
Abstract
Antigen-based rapid diagnostic tests (Ag-RDTs) were widely deployed to enhance SARS-CoV-2 testing capacity during the COVID-19 pandemic. Consistent with national guidance for low prevalence settings, positive Ag-RDTs were confirmed using nucleic acid amplification tests (NAATs) to avoid false positive results. However, increasing demands for positive Ag-RDT confirmation competed with other testing priorities in clinical laboratories. This work hypothesized that real-time RT-PCR without nucleic acid extraction (NAE) would be sufficiently sensitive to support positive Ag-RDT confirmation. Ag-RDT and NAAT results from community-based asymptomatic testing sites prior to the omicron variant wave were compared to calculate the weekly false positive rate (FPR) and false detection rate (FDR). Real-time RT-PCR was compared with and without NAE using 752 specimens previously tested positive for SARS-CoV-2 using commercial NAATs and 344 specimens from Ag-RDT-positive individuals. The impact of SARS-CoV-2 prevalence on laboratory resources required to sustain Ag-RDT confirmation was modeled for the RT-PCR with and without NAE. Overall, FPR was low [0.07% (222/330,763)] in asymptomatic testing sites, but FDR was high [30.7% (222/724)]. When RT-PCR was compared with and without NAE, 100% concordance was obtained with NAAT-positive specimens, including those from Ag-RDT-positive individuals. NAE-free RT-PCR significantly reduced time to results, human resources, and overall costs. A 30.7% FDR reaffirms the need for NAAT-based confirmation of positive Ag-RDT results during low SARS-CoV-2 prevalence. NAE-free RT-PCR was shown to be a simple and cost-sparing NAAT-based solution for positive Ag-RDT confirmation, and its implementation supported data-driven broader Ag-RDT deployment into communities, workplaces, and households. IMPORTANCE Rapid antigen testing for SARS-CoV-2 was widely deployed during the COVID-19 pandemic. In settings of low prevalence, national guidance recommends that positive antigen test results be confirmed with molecular testing. Given the high testing burden on clinical laboratories during the COVID-19 pandemic, the high volume of positive antigen tests submitted for confirmatory testing posed challenges for laboratory workflow. This study demonstrated that a simple PCR method without prior nucleic acid purification is an accurate and cost-effective solution for positive rapid antigen test confirmation. Implementing this method allowed molecular confirmatory testing for positive antigen tests to be sustained as antigen testing was expanded into large populations such as workplaces, schools, and households.
Collapse
Affiliation(s)
- Gregory R. McCracken
- Division of Microbiology, Department of Pathology and Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
| | - Glenn Patriquin
- Division of Microbiology, Department of Pathology and Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Todd F. Hatchette
- Division of Microbiology, Department of Pathology and Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ross J. Davidson
- Division of Microbiology, Department of Pathology and Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Barbara Goodall
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Lisa Barrett
- Division of Microbiology, Department of Pathology and Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - James MacDonald
- Division of Microbiology, Department of Pathology and Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
| | - Charles Heinstein
- Division of Microbiology, Department of Pathology and Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
| | - Janice Pettipas
- Nova Scotia Provincial Public Health Laboratory Network (PPHLN), Halifax, Nova Scotia, Canada
| | - John Ross
- Praxes Medical Group, Halifax, Nova Scotia, Canada
| | - Jason J. LeBlanc
- Division of Microbiology, Department of Pathology and Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
11
|
Graf S, Engelmann L, Jeleff Wölfler O, Albrecht I, Schloderer M, Kramer A, Klankermayer L, Gebhardt F, Chaker AM, Spinner CD, Schwab R, Wollenberg B, Protzer U, Hoffmann D. Reopening the Bavarian State Opera Safely: Hygiene Strategies and Incidence of COVID-19 in Artistic Staff During Theater Season 2020/2021. J Voice 2024; 38:798.e7-798.e20. [PMID: 34906415 PMCID: PMC8627642 DOI: 10.1016/j.jvoice.2021.11.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 01/16/2023]
Abstract
Due to the drastically rising coronavirus disease (COVID-19) incidence since March 2020, social life was shut down across the globe, and most opera houses were closed. As a result, there are limited data on SARS-CoV-2 infections among artists. The Bavarian State Opera has been reopened in September 2020. This study aimed to identify the incidence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection among employees in the Bavarian State Opera. In addition, the various hygiene strategies for the work groups within the institution are described. During the study period from September 1, 2020 to July 31, 2021, 10,061 nasopharyngeal swabs were obtained from 1,460 artistic staff members in a rolling system. During the entire study period, 61 individuals tested positive for SARS-CoV-2. None of the patients had a severe disease course. Compared to the seven-day-incidence per 100,000 German inhabitants, the estimated corresponding incidence among employees was lower at 37 weeks and higher or equal at 9 weeks. Among the infected individuals, 58.3% were symptomatic, 23.3% were presymptomatic, and 18.3% were asymptomatic. Forty-five percent of employees reported that they had been infected in their private environment, 41.7% suspected that their colleagues were the main contact, and 13.3% were unsure about the origin of their infection. Twenty-four diseased employees were ballet dancers, eight from the orchestra, seven from the administration, seven from the choir singers, six from the costume department, 10 from technical support, and one guest solo singer. In the 2020/2021 theater season, increased SARS-CoV-2 infections and large disease outbreaks were avoided at the Bavarian State Opera. Hygiene strategies, that existed since the beginning, was specifically designed for various work areas in the opera. Regular, mandatory PCR testing and follow-up of positive cases with the issuance of quarantine were performed. Using this disease management approach, artistic work at and reopening of the Bavarian State Opera was feasible with a well-controlled risk.
Collapse
Affiliation(s)
- Simone Graf
- Technical University of Munich, School of Medicine, Munich, University hospital rechts der Isar, Department of Otorhinolaryngology, Germany.
| | - Luca Engelmann
- Technical University of Munich, School of Medicine, Munich, University hospital rechts der Isar, Department of Otorhinolaryngology, Germany
| | - Olivia Jeleff Wölfler
- Technical University of Munich, School of Medicine, Munich, University hospital rechts der Isar, Department of Otorhinolaryngology, Germany
| | | | | | | | - Lucia Klankermayer
- Technical University of Munich, School of Medicine, Munich, University hospital rechts der Isar, Department of Otorhinolaryngology, Germany
| | - Friedemann Gebhardt
- Technical University of Munich /Helmholtz Center, School of Medicine, Munich, Institute for Medical Microbiology, Immunology and Hygiene, Germany
| | - Adam M Chaker
- Technical University of Munich, School of Medicine, Munich, University hospital rechts der Isar, Department of Otorhinolaryngology, Germany; Technical University of Munich, School of Medicine, Munich, University hospital rechts der Isar, Center of Allergy and Environment (ZAUM)
| | - Christoph D Spinner
- Technical University of Munich, School of Medicine, Munich, University hospital rechts der Isar, Department of Internal Medicine II, Germany
| | | | - Barbara Wollenberg
- Technical University of Munich, School of Medicine, Munich, University hospital rechts der Isar, Department of Otorhinolaryngology, Germany
| | - Ulrike Protzer
- Technical University of Munich /Helmholtz Center, School of Medicine, Munich, Institute of Virology, Germany
| | - Dieter Hoffmann
- Technical University of Munich /Helmholtz Center, School of Medicine, Munich, Institute of Virology, Germany
| |
Collapse
|
12
|
Wertenauer C, Dressel A, Wieland E, Wertenauer HJ, Braitmaier H, Straub A, Helfert S, Lützner N, März W. Diagnostic performance of rapid antigen testing for SARS-CoV-2: the COVid-19 AntiGen (COVAG) extension study. Front Med (Lausanne) 2024; 11:1352633. [PMID: 38698781 PMCID: PMC11064845 DOI: 10.3389/fmed.2024.1352633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/04/2024] [Indexed: 05/05/2024] Open
Abstract
Background This study is the extension of the COVAG study. We compared two RATs, the Panbio COVID-19 Ag Rapid Test (Abbott) and the SD Biosensor Q SARS-CoV-2 Rapid Antigen Test (Roche), against RT-PCR on the foil of new variants. Methods We included 888 all-comers at a diagnostic center between October 20, 2021, and March 18, 2022. RT-PCR-positive samples with a Ct value ≤32 were examined for SARS-CoV-2 variants. Findings The sensitivity of the Abbott-RAT and Roche-RAT were 65 and 67%, respectively. For both RATs, lower Ct values were significantly correlated with higher sensitivity. For samples with Ct values ≤25, the sensitivities of the Roche-RAT and of the Abbott-RAT were 96 and 95%, for Ct values 25-30 both were 19%, and for Ct values ≥30 they were 6 and 2%, respectively. The RATs had substantially higher sensitivities in symptomatic than asymptomatic participants (76, 77%, vs. 29, 31%, for Abbott-RAT, Roche-RAT, respectively) and in participants referred to testing by their primary care physician (84, 85%) compared to participants who sought testing due to referral by the health department (55, 58%) or a warning by the Corona-Warn-App (49, 49%). In persons with self-reported previous COVID-19 sensitivities were markedly lower than in patients without previous COVID-19: 27% vs. 75% for Roche-RAT and 27% vs. 73% for Abbott-RAT. We did not find significant correlation between vaccination status and sensitivity. The Omicron variant was detected with a sensitivity of 94 and 92%, the delta variant with a sensitivity of 80 and 80% for Abbott-RAT and Roche-RAT, respectively. This difference is attributable to the lower Ct values of the Omicron samples compared to the Delta samples. When adjusted for the Ct value, a multivariate logistic regression did not show a significant difference between Omicron and Delta. In terms of sensitivity, we found no significant difference between the wild-type and the Omicron and Delta variants, but a significantly lower sensitivity to the alpha variant compared to the other variants.The specificities were > 99% overall.
Collapse
Affiliation(s)
- Christoph Wertenauer
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Hausärzte am Schillerplatz, Stuttgart, Germany
- SYNLAB Holding Deutschland GmbH, Augsburg, Germany
| | | | - Eberhard Wieland
- SYNLAB Medical Care Center Leinfelden-Echterdingen GmbH, Leinfelden-Echterdingen, Germany
| | | | - Helmine Braitmaier
- SYNLAB Medical Care Center Leinfelden-Echterdingen GmbH, Leinfelden-Echterdingen, Germany
| | - Anna Straub
- SYNLAB Medical Care Center Leinfelden-Echterdingen GmbH, Leinfelden-Echterdingen, Germany
| | - Stefan Helfert
- SYNLAB MVZ Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Nicolas Lützner
- SYNLAB Medical Care Center Leinfelden-Echterdingen GmbH, Leinfelden-Echterdingen, Germany
| | - Winfried März
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| |
Collapse
|
13
|
Montero-Gómez A. [Diagnosis by isothermal amplification of nucleic acids. Opportunity for community pharmacies]. FARMACEUTICOS COMUNITARIOS 2024; 16:46-53. [PMID: 39156031 PMCID: PMC11328073 DOI: 10.33620/fc.2173-9218.(2024).14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/16/2024] [Indexed: 08/20/2024]
Abstract
This review focuses on describing new commercially available POC-type molecular diagnostic systems that can be easily implemented in community pharmacies and have the potential to expand the portfolio of pharmaceutical services and make a significant contribution to the improvement of public health.Knowledge of new molecular diagnostic techniques other than PCR is relatively unexplored. However, the available options are diverse and have reached sufficient technological maturity for large-scale use. The SARS-CoV-2 pandemic has brought diagnostic tests to market that, in some cases, have been used exclusively in research for decades.Isothermal nucleic acid amplification technology continues to evolve and it is likely that in the coming years we will witness an exponential increase in its use, as well as the development of new improvements that further simplify and reduce the cost of each assay.Furthermore, we cannot ignore the fact that during the COVID-19 pandemic, the public has become accustomed to self-diagnosing through mass distribution channels such as community pharmacies. Which can open the sector to other diseases - such as sexually transmitted diseases or animal health -, food control, water and air contamination (fungi) or the presence of allergens.Knowledge of them is an essential technological surveillance strategy for the pharmaceutical sector.
Collapse
|
14
|
Chen Y, Liu J, Shao S, Song Z, Ma Y, Tuo Y, Fang L, Xu Y, Xu B, Gu W, Cao X, Chen J, Yang Y, Wang P, Zhang J, Xu Y, Yu D, Hou P, Meng K, Li Z, Liu G, Qu X, Ji L, Yang R, Zhang L. Characteristics and outcomes of COVID-19 in Chinese immune thrombocytopenia patients: A prospective cohort study. Br J Haematol 2024; 204:1207-1218. [PMID: 37967471 DOI: 10.1111/bjh.19198] [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: 09/04/2023] [Revised: 10/09/2023] [Accepted: 10/30/2023] [Indexed: 11/17/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has a significant impact on the immune system. This is the first and largest study on pre-existing immune thrombocytopenia (ITP) patients infected with COVID-19 in China. We prospectively collected ITP patients infected with COVID-19 enrolled in the National Longitudinal Cohort of Hematological Diseases (NICHE, NCT04645199) and followed up for at least 1 month after infection. One thousand and one hundred forty-eight pre-existing ITP patients were included. Two hundred and twelve (18.5%) patients showed a decrease in the platelet (PLT) count after infection. Forty-seven (4.1%) patients were diagnosed with pneumonia. Risk factors for a decrease in the PLT count included baseline PLT count <50 × 109/L (OR, 1.76; 95% CI, 1.25-2.46; p = 0.001), maintenance therapy including thrombopoietin receptor agonists (TPO-RAs) (OR, 2.27; 95% CI, 1.60-3.21; p < 0.001) and previous splenectomy (OR, 1.98; 95% CI, 1.09-3.61; p = 0.03). Risk factors for pneumonia included age ≥40 years (OR, 2.45; 95% CI, 1.12-5.33; p = 0.02), ≥2 comorbidities (OR, 3.47; 95% CI, 1.63-7.64; p = 0.001), maintenance therapy including TPO-RAs (OR, 2.14; 95% CI, 1.17-3.91; p = 0.01) and immunosuppressants (OR, 3.05; 95% CI, 1.17-7.91; p = 0.02). In this cohort study, we described the characteristics of pre-existing ITP patients infected with COVID-19 and identified several factors associated with poor outcomes.
Collapse
Affiliation(s)
- Yunfei Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jiaying Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuai Shao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zhen Song
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yueshen Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yuanyuan Tuo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- Department of Pediatric Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lijun Fang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yanmei Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Bingqi Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wenjing Gu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xuan Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jia Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yu Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Panjing Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jing Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yuan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Dandan Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Pengxiao Hou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ke Meng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zhirong Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Guanyu Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xinmiao Qu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lulu Ji
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Renchi Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
15
|
Alsayed AR, Ahmed SI, AL Shweiki AO, Al-Shajlawi M, Hakooz N. The laboratory parameters in predicting the severity and death of COVID-19 patients: Future pandemic readiness strategies. BIOMOLECULES & BIOMEDICINE 2024; 24:238-255. [PMID: 37712883 PMCID: PMC10950347 DOI: 10.17305/bb.2023.9540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/30/2023] [Accepted: 09/14/2023] [Indexed: 09/16/2023]
Abstract
The range of clinical manifestations associated with the infection by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) encompasses a broad spectrum, ranging from flu-like symptoms to the occurrence of multiple organ failure and death. The severity of the coronavirus disease 2019 (COVID-19) is categorized based on clinical presentation and is divided into three distinct levels of severity identified as non-severe, severe, and critical. Although individuals of all age groups are susceptible to SARS-CoV-2 infection, middle-aged and older adults are more frequently impacted, with the latter being more likely to develop severe illness. Various laboratory characteristics observed in hospitalized COVID-19 patients have been correlated with adverse outcomes. These include elevated levels of D-dimer, liver enzymes, lactate dehydrogenase, C-reactive protein, ferritin, prothrombin time, and troponin, as well as decreased lymphocyte and platelets counts. This review investigated the relationship between baseline clinical characteristics, initial laboratory parameters upon hospital admission, and the severity of illness and mortality rates among COVID-19 patients. Although the COVID-19 pandemic has concluded, understanding the laboratory predictors of virus severity and mortality remains crucial, and examining these predictors can have long-term effects. Such insights can help healthcare systems manage resources more effectively and deliver timely and appropriate care by identifying and targeting high-risk individuals. This knowledge can also help us better prepare for future pandemics. By examining these predictors, we can take steps to protect public health and mitigate the impact of future pandemics.
Collapse
Affiliation(s)
- Ahmad R Alsayed
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Syed Imran Ahmed
- College of Health and Science, School of Pharmacy, University of Lincoln, Lincoln, United Kingdom
| | - Anas Osama AL Shweiki
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Mustafa Al-Shajlawi
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Nancy Hakooz
- School of Pharmacy, The University of Jordan, Amman, Jordan
| |
Collapse
|
16
|
Petersen A, Pienaar K. Competing realities, uncertain diagnoses of infectious disease: Mass self-testing for COVID-19 and liminal bio-citizenship. SOCIOLOGY OF HEALTH & ILLNESS 2024; 46:242-260. [PMID: 37526471 DOI: 10.1111/1467-9566.13694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 06/25/2023] [Indexed: 08/02/2023]
Abstract
Diagnoses of infectious diseases are being transformed as mass self-testing using rapid antigen tests (RATs) is increasingly integrated into public health. Widely used during the COVID-19 pandemic, RATs are claimed to have many advantages over 'gold-standard' polymerase chain reaction tests, especially their ease of use and production of quick results. Yet, while laboratory studies indicate the value of RATs in detecting the SARS-CoV-2 virus antigen, uncertainty surrounds their deployment and ultimate effectiveness in stemming infections. This article applies the analytic lens of biological citizenship (or bio-citizenship) to explore Australia's experience of implementing a RAT-based mass self-testing strategy to manage COVID-19. Drawing on Annemarie Mol's (1999, The Sociological Review, 47(1), 74-89) concept of ontological politics and analysing government statements, scientific articles and news media reporting published during a critical juncture of the strategy's implementation, we explore the kind of bio-citizenship implied by this strategy. Our analysis suggests the emergence of what we call liminal bio-citizenship, whereby citizens are made responsible for self-managing infection risk without the diagnostic certitude this demands. We discuss how the different realities of mass self-testing interact to reinforce this liminal citizenship and consider the implications for the sociology of diagnosis.
Collapse
Affiliation(s)
- Alan Petersen
- Sociology and Anthropology, School of Social Sciences, Faculty of Arts, Monash University, Melbourne, Victoria, Australia
| | - Kiran Pienaar
- Sociology, School of Humanities and Social Science, Faculty of Arts and Education, Deakin University, Melbourne, Victoria, Australia
| |
Collapse
|
17
|
Pannipulath Venugopal V, Babu Saheer L, Maktabdar Oghaz M. COVID-19 lateral flow test image classification using deep CNN and StyleGAN2. Front Artif Intell 2024; 6:1235204. [PMID: 38348096 PMCID: PMC10860423 DOI: 10.3389/frai.2023.1235204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 12/28/2023] [Indexed: 02/15/2024] Open
Abstract
Introduction Artificial intelligence (AI) in healthcare can enhance clinical workflows and diagnoses, particularly in large-scale operations like COVID-19 mass testing. This study presents a deep Convolutional Neural Network (CNN) model for automated COVID-19 RATD image classification. Methods To address the absence of a RATD image dataset, we crowdsourced 900 real-world images focusing on positive and negative cases. Rigorous data augmentation and StyleGAN2-ADA generated simulated images to overcome dataset limitations and class imbalances. Results The best CNN model achieved a 93% validation accuracy. Test accuracies were 88% for simulated datasets and 82% for real datasets. Augmenting simulated images during training did not significantly improve real-world test image performance but enhanced simulated test image performance. Discussion The findings of this study highlight the potential of the developed model in expediting COVID-19 testing processes and facilitating large-scale testing and tracking systems. The study also underscores the challenges in designing and developing such models, emphasizing the importance of addressing dataset limitations and class imbalances. Conclusion This research contributes to the deployment of large-scale testing and tracking systems, offering insights into the potential applications of AI in mitigating outbreaks similar to COVID-19. Future work could focus on refining the model and exploring its adaptability to other healthcare scenarios.
Collapse
Affiliation(s)
| | - Lakshmi Babu Saheer
- School of Computing and Information Science, Anglia Ruskin University, Cambridge, United Kingdom
| | | |
Collapse
|
18
|
Gobena D, Gudina EK, Gebre G, Degfie TT, Mekonnen Z. Rapid antigen test as a screening tool for SARS-CoV-2 infection: Head-to-head comparison with qRT-PCR in Ethiopia. Heliyon 2024; 10:e23518. [PMID: 38169801 PMCID: PMC10758869 DOI: 10.1016/j.heliyon.2023.e23518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Objective This study aimed to determine the diagnostic accuracy of the antigen rapid diagnostic test (Ag-RDT) as a screening tool for SARS-CoV-2 infection compared to Quantitative reverse transcription polymerase chain reaction (qRT-PCR). Methods This study was conducted at six referral hospitals in Oromia Region, Ethiopia. One thousand seven hundred twenty-one patients who visited the hospitals for various medical conditions were tested with qRT-PCR and/or Ag-RDTs. Qualitative detection of SARS-CoV-2 antigen was performed using the Panbio™ COVID-19 Ag rapid test device. Results Compared with qRT-PCR, Ag-RDTs had a sensitivity of 33.3 % (95%CI: 30.9%-35.9 %) and a specificity of 99.3 % (95%CI: 98.8%-99.7 %) to detect active SARS-CoV-2 infection. The area under the receiver operator curve was 0.67 (95%CI: 0.63-0.69). The sensitivity of Ag-RDTs appeared high in patients with shortness of breath (73.3 %) and those presenting with all three symptoms - fever, cough, and dyspnea (71.4 %). In all instances, specificity was more than 98 %. The Ag-RDT positivity rate also correlated well with viral load: 51.7 % in cases with cycle threshold (Ct) < 25 (high viral load) and only 3.4 % when Ct > 25 (low viral load). Conclusion Although Ag-RDT for diagnosing SARS-CoV-2 is a good option as a point-of-care screening tool, it has a low sensitivity to detect active infections. Using Panbio™ COVID-19 Ag Rapid test for diagnostic and treatment decisions may lead to a false negative, resulting in patient misdiagnosis, ultimately contributing to disease spread and poor patient outcome.
Collapse
Affiliation(s)
- Dabesa Gobena
- Public Health Emergency Management and Health Research Directorate, Oromia Health Bureau, Addis Ababa, Ethiopia
- School of Medical Laboratory Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
| | | | - Getu Gebre
- Public Health Emergency Management and Health Research Directorate, Oromia Health Bureau, Addis Ababa, Ethiopia
| | - Tizta Tilahun Degfie
- Department of Reproductive Health and Population Studies, College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
- Fenot Project, Department of Global Health and Population, Harvard T.H. Chan School, Ethiopia
| | - Zeleke Mekonnen
- School of Medical Laboratory Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
| |
Collapse
|
19
|
Katzenschlager S, Brümmer LE, Schmitz S, Tolle H, Manten K, Gaeddert M, Erdmann C, Lindner A, Tobian F, Grilli M, Pollock NR, Macé A, Erkosar B, Carmona S, Ongarello S, Johnson CC, Sacks JA, Denkinger CM, Yerlikaya S. Comparing SARS-CoV-2 antigen-detection rapid diagnostic tests for COVID-19 self-testing/self-sampling with molecular and professional-use tests: a systematic review and meta-analysis. Sci Rep 2023; 13:21913. [PMID: 38081881 PMCID: PMC10713601 DOI: 10.1038/s41598-023-48892-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Self-testing is an effective tool to bridge the testing gap for several infectious diseases; however, its performance in detecting SARS-CoV-2 using antigen-detection rapid diagnostic tests (Ag-RDTs) has not been systematically reviewed. This study aimed to inform WHO guidelines by evaluating the accuracy of COVID-19 self-testing and self-sampling coupled with professional Ag-RDT conduct and interpretation. Articles on this topic were searched until November 7th, 2022. Concordance between self-testing/self-sampling and fully professional-use Ag-RDTs was assessed using Cohen's kappa. Bivariate meta-analysis yielded pooled performance estimates. Quality and certainty of evidence were evaluated using QUADAS-2 and GRADE tools. Among 43 studies included, twelve reported on self-testing, and 31 assessed self-sampling only. Around 49.6% showed low risk of bias. Overall concordance with professional-use Ag-RDTs was high (kappa 0.91 [95% confidence interval (CI) 0.88-0.94]). Comparing self-testing/self-sampling to molecular testing, the pooled sensitivity and specificity were 70.5% (95% CI 64.3-76.0) and 99.4% (95% CI 99.1-99.6), respectively. Higher sensitivity (i.e., 93.6% [95% CI 90.4-96.8] for Ct < 25) was estimated in subgroups with higher viral loads using Ct values as a proxy. Despite high heterogeneity among studies, COVID-19 self-testing/self-sampling exhibits high concordance with professional-use Ag-RDTs. This suggests that self-testing/self-sampling can be offered as part of COVID-19 testing strategies.Trial registration: PROSPERO: CRD42021250706.
Collapse
Affiliation(s)
- Stephan Katzenschlager
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Lukas E Brümmer
- Division of Infectious Disease and Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg University Hospital, Heidelberg, Germany
| | - Stephani Schmitz
- Division of Infectious Disease and Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
- Department of Developmental Biology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hannah Tolle
- Division of Infectious Disease and Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Katharina Manten
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- Division of Infectious Disease and Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Mary Gaeddert
- Division of Infectious Disease and Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | | | - Andreas Lindner
- Charité Center for Global Health, Institute of International Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tobian
- Division of Infectious Disease and Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | | | - Nira R Pollock
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA, USA
| | | | | | | | | | - Cheryl C Johnson
- Global HIV, Hepatitis and STIs Programmes, World Health Organization, Geneva, Switzerland
| | - Jilian A Sacks
- Department of Epidemic and Pandemic Preparedness and Prevention, World Health Organization, Geneva, Switzerland
| | - Claudia M Denkinger
- Division of Infectious Disease and Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg University Hospital, Heidelberg, Germany
| | - Seda Yerlikaya
- Division of Infectious Disease and Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.
| |
Collapse
|
20
|
Wang Q, Gu H, Tao Y, Zhao Y, Meng Z. Number of initial symptoms of SARS-CoV-2 infection is associated with the risk of otological symptoms: a retrospective study. BMC Infect Dis 2023; 23:862. [PMID: 38062350 PMCID: PMC10704705 DOI: 10.1186/s12879-023-08866-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/04/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The characteristics of otological symptoms in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are lacking. Almost no research has been conducted to explore the emergence of otological symptoms after coronavirus disease 2019 infection. The aims of this study were to investigate the prevalence and specific clinical characteristics of and risk factors for otological symptoms among patients with SARS-CoV-2 infection. METHODS We included two groups to investigate the prevalence and clinical characteristics of otological symptoms among patients with SARS-CoV-2 infection. The first sample (S1) was drawn retrospectively from four communities via questionnaires, and the second sample (S2) from an outpatient clinic. RESULTS A total of 189 participants were included in S1 (124 women [65.6%]; mean [standard deviation (SD)] age, 33.66 [13.56] years), and 47 in S2 (25 women [53.2%]; mean [SD] age, 45.28 [14.64] years). The most prevalent otological symptoms in S1 were dizziness (15.9%), tinnitus (7.9%), aural fullness (6.9%), otalgia (5.3%), hearing loss (1.6%), and otopyorrhoea (1.1%). Moreover, for each additional typical symptom of SARS-CoV-2 infection, the risk (odds ratio) of otological symptoms increased by 1.33 (95% confidence interval: 1.10-1.61, p = 0.003). The prevalence of aural fullness was higher in the unvaccinated group than that in the group receiving two or three vaccinations (p = 0.018). CONCLUSIONS Various otological symptoms may occur in patients with SARS-CoV-2 infection. The number of typical symptoms of SARS-CoV-2 infection is positively associated with the probability of otological symptoms. However, vaccination may reduce the probability of certain otological symptoms.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital, Sichuan University, 37 Guo Xue Lane, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Audiology and Speech Language Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Hailing Gu
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital, Sichuan University, 37 Guo Xue Lane, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Audiology and Speech Language Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Tao
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital, Sichuan University, 37 Guo Xue Lane, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Audiology and Speech Language Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Zhao
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital, Sichuan University, 37 Guo Xue Lane, Chengdu, 610041, Sichuan, People's Republic of China.
- Department of Audiology and Speech Language Pathology, West China Hospital, Sichuan University, Chengdu, China.
| | - Zhaoli Meng
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital, Sichuan University, 37 Guo Xue Lane, Chengdu, 610041, Sichuan, People's Republic of China.
- Department of Audiology and Speech Language Pathology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
21
|
García-Fernández S, Fernández-Morán E, López-Martínez C, Vivanco-Allende B, Costales-Álvarez C, Ordóñez-Álvarez FA. Tubulointerstitial nephritis and uveitis syndrome and SARS-CoV-2 infection in an adolescent: just a coincidence in time? Pediatr Nephrol 2023; 38:4203-4207. [PMID: 37130974 PMCID: PMC10153047 DOI: 10.1007/s00467-023-05950-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND Despite recent well-established kidney tropism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), usually presenting as acute kidney injury (AKI), there are few published cases with SARS-CoV-2-related tubulointerstitial nephritis (TIN). We report an adolescent with TIN and delayed association with uveitis (TINU syndrome), where SARS-CoV-2 spike protein was identified in kidney biopsy. CASE-DIAGNOSIS/TREATMENT A 12-year-old girl was assessed for a mild elevation of serum creatinine detected during the evaluation of systemic manifestations including asthenia, anorexia, abdominal pain, vomiting, and weight loss. Data of incomplete proximal tubular dysfunction (hypophosphatemia and hypouricemia with inappropriate urinary losses, low molecular weight proteinuria, and glucosuria) were also associated. Symptoms had initiated after a febrile respiratory infection with no known infectious cause. After 8 weeks, the patient tested positive in PCR for SARS-CoV-2 (Omicron variant). A subsequent percutaneous kidney biopsy revealed TIN and immunofluorescence staining with confocal microscopy detected the presence of SARS-CoV-2 protein S within the kidney interstitium. Steroid therapy was started with gradual tapering. Ten months after onset of clinical manifestations, as serum creatinine remained slightly elevated and kidney ultrasound showed mild bilateral parenchymal cortical thinning, a second percutaneous kidney biopsy was performed, without demonstrating acute inflammation or chronic changes, but SARS-CoV-2 protein S within the kidney tissue was again detected. At that moment, simultaneous routine ophthalmological examination revealed an asymptomatic bilateral anterior uveitis. CONCLUSIONS We present a patient who was found to have SARS-CoV-2 in kidney tissue several weeks following onset of TINU syndrome. Although simultaneous infection by SARS-CoV-2 could not be demonstrated at onset of symptoms, since no other etiological cause was identified, we hypothesize that SARS-CoV-2 might have been involved in triggering the patient's illness.
Collapse
Affiliation(s)
| | - Eva Fernández-Morán
- Department of Pediatrics, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Cecilia López-Martínez
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Centro de Investigación Biomédica En Red–Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Blanca Vivanco-Allende
- Department of Pathology, Hospital Universitario Central de Asturias, Oviedo, Spain
- Departamento de Cirugía y Especialidades Médico-Quirúrgicas, Área de Anatomía Patológica, Universidad de Oviedo, Oviedo, Spain
| | | | | |
Collapse
|
22
|
Chen Y, Feng L, Han Y, Zhao Z, Diao Z, Huang T, Ma Y, Feng W, Li J, Li Z, Liu C, Chang L, Li J, Zhang R. Performance evaluation of SARS-CoV-2 antigen detection in the post-pandemic era: multi-laboratory assessment. Clin Chem Lab Med 2023; 61:2237-2247. [PMID: 37377068 DOI: 10.1515/cclm-2023-0597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023]
Abstract
OBJECTIVES Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antigen detection is an indispensable tool for epidemic surveillance in the post-pandemic era. Faced with irregular performance, a comprehensive external quality assessment (EQA) scheme was conducted by the National Center for Clinical Laboratories (NCCL) to evaluate the analytical performance and status of SARS-CoV-2 antigen tests. METHODS The EQA panel included ten lyophilized samples containing serial 5-fold dilutions of inactivated SARS-CoV-2-positive supernatants of the Omicron BA.1 and BA.5 strains and negative samples, which were classified into "validating" samples and "educational" samples. Data were analyzed according to qualitative results for each sample. RESULTS A total of 339 laboratories in China participated in this EQA scheme, and 378 effective results were collected. All validating samples were correctly reported by 90.56 % (307/339) of the participants and 90.21 % (341/378) of the datasets. The positive percent agreement (PPA) was >99 % for samples with concentrations of 2 × 107 copies/mL but was 92.20 % (697/756) for 4 × 106 copies/mL and 25.26 % (382/1,512) for 8 × 105 copies/mL samples. Colloidal gold was the most frequently used (84.66 %, 320/378) but showed the lowest PPAs (57.11 %, 1,462/2,560) for positive samples compared with fluorescence immunochromatography (90 %, 36/40) and latex chromatography (79.01 %, 335/424). Among 11 assays used in more than 10 clinical laboratories, ACON showed a higher sensitivity than other assays. CONCLUSIONS The EQA study can help to validate whether it's necessary to update antigen detection assays for manufacturers and provide participants with information about the performance of assays to take the first step toward routine post-market surveillance.
Collapse
Affiliation(s)
- Yuqing Chen
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Lei Feng
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Yanxi Han
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Zihong Zhao
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
- Peking University Fifth School of Clinical Medicine, Beijing, P.R. China
| | - Zhenli Diao
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Tao Huang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Yu Ma
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Wanyu Feng
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Jing Li
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Ziqiang Li
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Cong Liu
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Lu Chang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Jinming Li
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
- National Center for Clinical Laboratories, Beijing Hospital, Beijing, P.R. China
| | - Rui Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
- National Center for Clinical Laboratories, Beijing Hospital, Beijing, P.R. China
| |
Collapse
|
23
|
Heyne TF, Negishi K, Choi DS, Al Saud AA, Marinacci LX, Smithedajkul PY, Devaraj LR, Little BP, Mendoza DP, Flores EJ, Petranovic M, Toal SP, Shokoohi H, Liteplo AS, Geisler BP. Handheld Lung Ultrasound to Detect COVID-19 Pneumonia in Inpatients: A Prospective Cohort Study. POCUS JOURNAL 2023; 8:175-183. [PMID: 38099168 PMCID: PMC10721309 DOI: 10.24908/pocus.v8i2.16484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Background: Chest imaging, including chest X-ray (CXR) and computed tomography (CT), can be a helpful adjunct to nucleic acid test (NAT) in the diagnosis and management of Coronavirus Disease 2019 (COVID-19). Lung point of care ultrasound (POCUS), particularly with handheld devices, is an imaging alternative that is rapid, highly portable, and more accessible in low-resource settings. A standardized POCUS scanning protocol has been proposed to assess the severity of COVID-19 pneumonia, but it has not been sufficiently validated to assess diagnostic accuracy for COVID-19 pneumonia. Purpose: To assess the diagnostic performance of a standardized lung POCUS protocol using a handheld POCUS device to detect patients with either a positive NAT or a COVID-19-typical pattern on CT scan. Methods: Adult inpatients with confirmed or suspected COVID-19 and a recent CT were recruited from April to July 2020. Twelve lung zones were scanned with a handheld POCUS machine. Images were reviewed independently by blinded experts and scored according to the proposed protocol. Patients were divided into low, intermediate, and high suspicion based on their POCUS score. Results: Of 79 subjects, 26.6% had a positive NAT and 31.6% had a typical CT pattern. The receiver operator curve for POCUS had an area under the curve (AUC) of 0.787 for positive NAT and 0.820 for a typical CT. Using a two-point cutoff system, POCUS had a sensitivity of 0.90 and 1.00 compared to NAT and typical CT pattern, respectively, at the lower cutoff; it had a specificity of 0.90 and 0.89 compared to NAT and typical CT pattern at the higher cutoff, respectively. Conclusions: The proposed lung POCUS protocol with a handheld device showed reasonable diagnostic performance to detect inpatients with a positive NAT or typical CT pattern for COVID-19. Particularly in low-resource settings, POCUS with handheld devices may serve as a helpful adjunct for persons under investigation for COVID-19 pneumonia.
Collapse
Affiliation(s)
- Thomas F Heyne
- Department of Medicine, Massachusetts General HospitalBoston, MAUSA
- Department of Pediatrics, Massachusetts General HospitalBoston, MAUSA
| | - Kay Negishi
- Department of Medicine, Massachusetts General HospitalBoston, MAUSA
| | - Daniel S Choi
- Department of Emergency Medicine, Massachusetts General HospitalBoston, MAUSA
| | - Ahad A Al Saud
- Department of Emergency Medicine, Massachusetts General HospitalBoston, MAUSA
- Department of Emergency Medicine, King Saud University College of MedicineRiyadhSaudi Arabia
| | - Lucas X Marinacci
- Richard A. and Susan F. Smith Center for Outcomes Research, Beth Israel Deaconess Medical CenterBoston, MAUSA
| | | | - Lily R Devaraj
- Department of Medicine, Massachusetts General HospitalBoston, MAUSA
- Department of Pediatrics, Massachusetts General HospitalBoston, MAUSA
| | - Brent P Little
- Department of Radiology, Massachusetts General HospitalBoston, MAUSA
| | - Dexter P Mendoza
- Department of Radiology, Massachusetts General HospitalBoston, MAUSA
| | - Efren J Flores
- Department of Radiology, Massachusetts General HospitalBoston, MAUSA
| | - Milena Petranovic
- Department of Radiology, Massachusetts General HospitalBoston, MAUSA
| | - Steven P Toal
- Department of Medicine, Massachusetts General HospitalBoston, MAUSA
| | - Hamid Shokoohi
- Department of Emergency Medicine, Massachusetts General HospitalBoston, MAUSA
| | - Andrew S Liteplo
- Department of Emergency Medicine, Massachusetts General HospitalBoston, MAUSA
| | - Benjamin P Geisler
- Department of Medicine, Massachusetts General HospitalBoston, MAUSA
- Institute for Medical Information Processing, Biometry, and Epidemiology, Ludwig Maximilian UniversityMunichGermany
| |
Collapse
|
24
|
Kinne V, Ehrenberg S, Baier M, Lang S, Lewejohann JC, Kipp F. [Diagnostic performance of two antigen-tests compared to one PCR-Test to detect SARS-CoV-2 in an emergency department and emergency service]. DIE ANAESTHESIOLOGIE 2023; 72:791-798. [PMID: 37792046 PMCID: PMC10615947 DOI: 10.1007/s00101-023-01343-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/04/2023] [Accepted: 08/27/2023] [Indexed: 10/05/2023]
Abstract
BACKGROUND In the German hospital landscape and emergency care the COVID-19 pandemic was a stress test. Emergency medical health care in Germany is ensured by the supply chain between prehospital emergency rescue and clinical emergency care in the emergency rooms. In hospitals and emergency care settings a rapid, simple, accurate, and cost-effective test is needed to identify SARS-CoV‑2. In the central emergency department it is important to strictly separate patients with suspected COVID-19 from non-infected emergency persons. METHODS Given the background mentioned above, the performance of antigen tests in the ambulance service of the city Jena and the central emergency department of the university hospital Jena was analysed and in addition verified by using the RT-PCR gold standard. Several multiple testing procedures were performed by using antigen tests in the ambulance service and the central emergency department, and by using one or both of these antigen tests followed by the RT-PCR test. A total of 980 patients were included in the study over a two-month period (October/November 2022). RESULTS The average age of all patients was 65 years. More than half of the actively treated patients came from the city of Jena. The sensitivity and specificity of the antigen tests were 66.7% and 99.2% in the clinical setting (the central emergency department) and 68.8% and 96.7% in the prehospital setting (in the ambulance service) compared to RT-PCR. In the prehospital setting the sensitivity of the antigen testing was slightly higher (2%) than the clinical antigen testing. Regarding the parallel testing, 6% of antigen tests had a false negative SARS-CoV‑2 antigen test result in the ambulance service and 4.6% of antigen tests had a false negative SARS-CoV‑2 antigen test result in the central emergency department. The false-negative antigen tests, and thus the potentially unrecognized individuals, were further reviewed by considering the Ct-value. CONCLUSION The use of antigen testing in the ambulance service and the emergency department can lead to a quick classification of COVID and non-COVID areas of an emergency department. The measurement accuracy of antigen testing in the ambulance service and central emergency department is not equivalent to the RT-PCR. Nevertheless, antigen testing is a useful initial screening tool for early detection of SARS-CoV‑2 in prehospital and clinical settings. Dual antigen testing may be useful for more accurate diagnosis of the SARS-CoV‑2 pathogen.
Collapse
Affiliation(s)
- Veit Kinne
- Institut für Infektionsmedizin und Krankenhaushygiene, Universitätsklinikum Jena, Jena, Deutschland.
| | - Sandra Ehrenberg
- Institut für Infektionsmedizin und Krankenhaushygiene, Universitätsklinikum Jena, Jena, Deutschland
| | - Michael Baier
- Institut für Medizinische Mikrobiologie, Universitätsklinikum Jena, Jena, Deutschland
| | - Sebastian Lang
- Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Jena, Jena, Deutschland
| | | | - Frank Kipp
- Institut für Infektionsmedizin und Krankenhaushygiene, Universitätsklinikum Jena, Jena, Deutschland
| |
Collapse
|
25
|
Ayran J, Köhler C, Linh LTK, Schneider G, Pallerla SR, Battke F, Federle L, Martus P, Kremsner PG, Velavan TP. Tübingen model study: large-scale introduction of rapid antigen testing in the population and the viral dynamics of SARS-CoV-2. Front Public Health 2023; 11:1159622. [PMID: 37942246 PMCID: PMC10628735 DOI: 10.3389/fpubh.2023.1159622] [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: 02/06/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023] Open
Abstract
Despite of contact restrictions, population mobility remains the main reason for the spread of SARS-CoV-2. The state of Baden-Württemberg (BW), Germany, approved a model study in Tübingen (TÜMOD) to evaluate how mandatory rapid diagnostic tests (RDT) could reduce transmission. Between 16 March and 24 April 2021, approximately 165,000 residents and visitors to the city were screened for SARS CoV-2 infection using Abbott Panbio™ COVID-19 Antigen rapid test device. We assessed incidences and recorded epidemiological characteristics in a subset of 4,118 participants recruited at three of the nine testing stations. PCR tests were performed in RDT-positives to determine the positive predictive value (PPV), and circulating variants of SARS-CoV-2 were identified by whole-genome sequencing. 2,282 RDT-negative samples were tested by pooled PCR to calculate the false negative rate (FNR). Viral load was compared between variants. 116 (3%) participants were positive by RDT, and of these, 57 (49%) were positive by PCR, 55 (47%) were negative. This resulted in a PPV of 51%. Of the 57 positives, 52 SARS-CoV-2 genomes were successfully sequenced. Of these, 50 belonged to the B.1.1.7 lineage, which had a high viral load (average Ct = 19). Of the 2,282 RDT negatives tested, all were PCR negative (FNR 0%). At the end of TÜMOD, the incidence in Tübingen, which was initially lower, had reached the incidence in the state of BW. While it is difficult to assess the impact of TÜMOD on incidence independent of confounding factors, further studies are needed to identify the effect of close-meshed testing on infection rates.
Collapse
Affiliation(s)
- Jule Ayran
- Institute of Tropical Medicine, Travel Medicine and Human Parasitology, Competence Centre for Tropical Medicine Baden-Württemberg, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Carsten Köhler
- Institute of Tropical Medicine, Travel Medicine and Human Parasitology, Competence Centre for Tropical Medicine Baden-Württemberg, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Le Thi Kieu Linh
- Institute of Tropical Medicine, Travel Medicine and Human Parasitology, Competence Centre for Tropical Medicine Baden-Württemberg, Universitätsklinikum Tübingen, Tübingen, Germany
- Vietnamese-German Center for Medical Research, VG-CARE, Hanoi, Vietnam
| | - Gisela Schneider
- DIFAEM – German Institute for Medical Mission, Tübingen, Germany
| | - Srinivas Reddy Pallerla
- Institute of Tropical Medicine, Travel Medicine and Human Parasitology, Competence Centre for Tropical Medicine Baden-Württemberg, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Florian Battke
- Center for Genomics and Transcriptomics (CeGaT) GmbH, Tübingen, Germany
| | - Lisa Federle
- German Red Cross, Regional Centre Tübingen, Tübingen, Germany
| | - Peter Martus
- Institute for Clinical Epidemiology and Applied Biometry, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Peter G. Kremsner
- Institute of Tropical Medicine, Travel Medicine and Human Parasitology, Competence Centre for Tropical Medicine Baden-Württemberg, Universitätsklinikum Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambarene, Gabon
| | - Thirumalaisamy P. Velavan
- Institute of Tropical Medicine, Travel Medicine and Human Parasitology, Competence Centre for Tropical Medicine Baden-Württemberg, Universitätsklinikum Tübingen, Tübingen, Germany
- Vietnamese-German Center for Medical Research, VG-CARE, Hanoi, Vietnam
- Faculty of Medicine, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
26
|
Abell I, Zachreson C, Conway E, Geard N, McVernon J, Waring T, Baker C. Rapid prototyping of models for COVID-19 outbreak detection in workplaces. BMC Infect Dis 2023; 23:713. [PMID: 37872480 PMCID: PMC10591376 DOI: 10.1186/s12879-023-08713-y] [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: 03/06/2023] [Accepted: 10/15/2023] [Indexed: 10/25/2023] Open
Abstract
Early case detection is critical to preventing onward transmission of COVID-19 by enabling prompt isolation of index infections, and identification and quarantining of contacts. Timeliness and completeness of ascertainment depend on the surveillance strategy employed. This paper presents modelling used to inform workplace testing strategies for the Australian government in early 2021. We use rapid prototype modelling to quickly investigate the effectiveness of testing strategies to aid decision making. Models are developed with a focus on providing relevant results to policy makers, and these models are continually updated and improved as new questions are posed. Developed to support the implementation of testing strategies in high risk workplace settings in Australia, our modelling explores the effects of test frequency and sensitivity on outbreak detection. We start with an exponential growth model, which demonstrates how outbreak detection changes depending on growth rate, test frequency and sensitivity. From the exponential model, we learn that low sensitivity tests can produce high probabilities of detection when testing occurs frequently. We then develop a more complex Agent Based Model, which was used to test the robustness of the results from the exponential model, and extend it to include intermittent workplace scheduling. These models help our fundamental understanding of disease detectability through routine surveillance in workplaces and evaluate the impact of testing strategies and workplace characteristics on the effectiveness of surveillance. This analysis highlights the risks of particular work patterns while also identifying key testing strategies to best improve outbreak detection in high risk workplaces.
Collapse
Affiliation(s)
- Isobel Abell
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, Australia.
- Melbourne Centre for Data Science, The University of Melbourne, Melbourne, Australia.
| | - Cameron Zachreson
- School of Computing and Information Systems, The University of Melbourne, Melbourne, Australia
| | - Eamon Conway
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Nicholas Geard
- School of Computing and Information Systems, The University of Melbourne, Melbourne, Australia
| | - Jodie McVernon
- Peter Doherty Institute for Infection and Immunity, The University of Melbourne and the Royal Melbourne Hospital, Melbourne, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, Australia
| | - Thomas Waring
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, Australia
- Melbourne Centre for Data Science, The University of Melbourne, Melbourne, Australia
- Centre of Excellence for Biosecurity Risk Analysis, The University of Melbourne, Melbourne, Australia
| | - Christopher Baker
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, Australia
- Melbourne Centre for Data Science, The University of Melbourne, Melbourne, Australia
- Centre of Excellence for Biosecurity Risk Analysis, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
27
|
Okude M, Suzuki K, Naito A, Ebashi A, Kusama T, Kiyotaki J, Akashi Y, Kiyasu Y, Kurihara Y, Notake S, Takanashi M, Setoyama T, Kawakami Y, Suzuki H. Development of a mobile laboratory system in hydrogen fuel cell buses and evaluation of the performance for COVID-19 RT-PCR testing. Sci Rep 2023; 13:17546. [PMID: 37845364 PMCID: PMC10579409 DOI: 10.1038/s41598-023-44925-7] [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: 01/30/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023] Open
Abstract
We designed and developed two new types of hydrogen fuel cell (HFC) buses (motorcoach and minibus) with a mobile laboratory system. Feasibility studies have been performed for mobile laboratory testing, particularly for the laboratory performance of COVID-19 RT-PCR (PCR). We evaluated the driving range capability, PCR sample size capacity, turnaround time (TAT), and analytical performance for the detection of SARS-CoV-2. Saliva samples were used for the current study, and the analytical performance was compared with that of the reference PCR. The estimated driving range and sample size capacity of the HFC and HFC minibus were 432 km and 2847 samples, respectively, for the HFC motorcoach and 313 km and 1949 samples for the HFC minibus. For the TAT, the median time between sample submission and completion of PCR was 86 min for the motorcoach and 76 min for the minibus, and the median time between sample submission and electronic reporting of the result to each visitor was 182 min for the motorcoach and 194 min for the minibus. A secondary analysis of 1574 HFC mobile laboratory testing samples was conducted, and all negative samples were found to be negative by reference PCR. Furthermore, all samples were confirmed to be positive by reference PCR or other molecular examinations.
Collapse
Affiliation(s)
- Miho Okude
- LSI Medience Corporation, 3-30-1 Shimura, Itabashi, Tokyo, 174-8555, Japan
| | - Kenji Suzuki
- Center for Cybernics Research, University of Tsukuba, 1-1-1 Tennodai, TsukubaTsukuba, Ibaraki, 305-8573, Japan
| | - Asami Naito
- LSI Medience Corporation, 3-30-1 Shimura, Itabashi, Tokyo, 174-8555, Japan
| | - Akio Ebashi
- Department of Infectious Diseases, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 305-8576, Japan
| | - Tomoka Kusama
- Department of Infectious Diseases, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 305-8576, Japan
| | - Junichi Kiyotaki
- Miroku Medical Laboratory Inc, 659-2 Innai, Saku, Nagano, 384-2201, Japan
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 305-8558, Japan
| | - Yusaku Akashi
- Department of Infectious Diseases, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yoshihiko Kiyasu
- Department of Infectious Diseases, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 305-8576, Japan
- Department of Infectious Diseases, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
- Division of Infectious Diseases, Department of Medicine, Tsukuba Medical Center Hospital, 1-3-1 Amakubo Tsukuba, Ibaraki, 305-8558, Japan
| | - Yoko Kurihara
- Department of Infectious Diseases, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 305-8576, Japan
| | - Shigeyuki Notake
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 305-8558, Japan
| | - Masaki Takanashi
- LSI Medience Corporation, 3-30-1 Shimura, Itabashi, Tokyo, 174-8555, Japan
| | - Tomokazu Setoyama
- LSI Medience Corporation, 3-30-1 Shimura, Itabashi, Tokyo, 174-8555, Japan
| | - Yasushi Kawakami
- Department of Laboratory Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hiromichi Suzuki
- Department of Infectious Diseases, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 305-8576, Japan.
- Department of Infectious Diseases, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
28
|
Rebbapragada A, Cariazo L, Elchuk D, Abdelrahman H, Pham D, Raveendraraj J, Chokshi K, Joseph N, Gouzenkova E, Gill H, Blecher P. Performance of the Cue COVID-19 point-of-care molecular test: insights from a multi-site clinic service model. Microbiol Spectr 2023; 11:e0406422. [PMID: 37728337 PMCID: PMC10580901 DOI: 10.1128/spectrum.04064-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 07/26/2023] [Indexed: 09/21/2023] Open
Abstract
The COVID-19 pandemic highlighted the critical need for rapid and accurate molecular diagnostic testing. The Cue COVID-19 Point-of-Care Test (Cue POCT) is a nucleic acid amplification test (NAAT), authorized by Health Canada and FDA as a POCT for SARS-CoV-2 detection. Cue POCT was deployed at a network of clinics in Ontario, Canada with n = 13,848 patrons tested between 17 July 2021 and 31 January 2022. The clinical performance and operational experience with Cue POCT were examined for this testing population composed mostly of asymptomatic individuals (93.7%). A head-to-head prospective clinical verification was performed between 17 July and 4 October for all POCT service clients (n = 3,037) with paired COVID-19 testing by Cue and RT-PCR. Prospective verification demonstrated a clinical sensitivity of 100% and clinical specificity of 99.4% for Cue COVID-19 POCT. The lack of false negatives and low false positive rate (0.64%), underscores the high accuracy (99.4%) of Cue POCT to provide rapid PCR quality results. Low error rates (cancellation rate of 0% and invalid rate of 0.63%) with the current software version were additionally noted. Taken together, these findings highlight the value of accurate molecular COVID-19 POCT in a distributed service delivery model to rapidly detect cases in the community with the potential to curb transmission in high-exposure settings (i.e., in-flight, congregate workplace, and social events). The insights gleaned from this operational implementation are readily transferable to future POCT diagnostic services. IMPORTANCE This manuscript reports on the findings of a large asymptomatic population who underwent surveillance COVID testing on the Cue COVID-19 Point-of-Care Test (POCT). Review of test performance of this rapid molecular POCT, as compared to gold standard RT-PCR, is valuable to many audiences, including public health, emergency testing services, employers, and the general population of consumers who are seeking a user-friendly, accurate, cost-effective, and sustainable testing model for COVID screening. The findings from this operational experience also inform future models of POCT services beyond COVID.
Collapse
|
29
|
Oloo MA, Awandu SS, Onyango B, Magwanga RO, Oluoch AO, Lidechi S, Muok EM, Munga S, Estambale B. Comparative analysis of SARS-CoV-2 detection methods using stool, blood, and nasopharyngeal swab samples. Pan Afr Med J 2023; 46:21. [PMID: 38107343 PMCID: PMC10724033 DOI: 10.11604/pamj.2023.46.21.39483] [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: 03/01/2023] [Accepted: 09/11/2023] [Indexed: 12/19/2023] Open
Abstract
Introduction as a public health policy, the ongoing global coronavirus disease 2019 vaccination drives require continuous tracking, tracing, and testing of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Diagnostic testing is important in virus detection and understanding its spread for timely intervention. This is especially important for low-income settings where the majority of the population remains untested. This is well supported by the fact that of about 9% of the Kenyan population had been tested for the virus. Methods this was a cross-sectional study conducted at the Kisumu and Siaya Referral Hospitals in Kenya. Here we report on the sensitivity and specificity of the rapid antigen detection test (Ag-RDT) of SARS-CoV-2 compared with the quantitative reverse transcriptase polymerase chain reaction (RT-qPCR) using stool and nasopharyngeal swab samples. Further, the mean Immunoglobulin M (IgM) and Immunoglobulin G (IgG) antibody levels among symptomatic and asymptomatic individuals in western Kenya were evaluated. Results the sensitivity and specificity of Ag-RDT were 76.3% (95% CI, 59.8-88.6%) and 96.3% (95% CI, 87.3-99.5%) with a negative and positive predictive value of 85% (95% CI, 73.8%-93.0%) and 93% (95% CI, 78.6%-99.2%) respectively. There was substantial agreement of 88% (Kappa value of 0.75, 95% CI, 0.74-0.77) between Ag-RDT and nasopharyngeal swab RT-qPCR, and between stool and nasopharyngeal swab RT-qPCR results (83.7% agreement, Kapa value 0.62, 95% CI 0.45-0.80). The mean IgM and IgG antibody response to SARS-CoV-2 were not different in asymptomatic individuals, 1.11 (95% CI, 0.78-1.44) and 0.88 (95% CI, 0.65-1.11) compared to symptomatic individuals 4.30 (95% CI 3.30-5.31) and 4.16 (95% CI 3.32 -5.00). Conclusion the choice of an appropriate SARS-CoV-2 diagnostic, screening, and surveillance test should be guided by the specific study needs and a rational approach for optimal results.
Collapse
Affiliation(s)
- Marceline Adhiambo Oloo
- School of Biological, Physical, Mathematics and Actuarial Sciences, Jaramogi Oginga Odinga University of Science and Technology, P.O Box 210-40601, Bondo, Kenya
| | - Shehu Shagari Awandu
- School of Health Sciences, Jaramogi Oginga Odinga University of Science and Technology, P.O Box 210-40601, Bondo, Kenya
| | - Benson Onyango
- School of Biological, Physical, Mathematics and Actuarial Sciences, Jaramogi Oginga Odinga University of Science and Technology, P.O Box 210-40601, Bondo, Kenya
| | - Richard Odongo Magwanga
- School of Biological, Physical, Mathematics and Actuarial Sciences, Jaramogi Oginga Odinga University of Science and Technology, P.O Box 210-40601, Bondo, Kenya
| | - Alfred Ochieng Oluoch
- School of Biological, Physical, Mathematics and Actuarial Sciences, Jaramogi Oginga Odinga University of Science and Technology, P.O Box 210-40601, Bondo, Kenya
| | - Shirley Lidechi
- Kenya Medical Research Institute Centre for Global Health Research (CGHR), P.O Box 1578-40100, Kisumu, Kenya
| | - Erick Mbata Muok
- Kenya Medical Research Institute Centre for Global Health Research (CGHR), P.O Box 1578-40100, Kisumu, Kenya
| | - Stephen Munga
- Kenya Medical Research Institute Centre for Global Health Research (CGHR), P.O Box 1578-40100, Kisumu, Kenya
| | - Benson Estambale
- School of Health Sciences, Jaramogi Oginga Odinga University of Science and Technology, P.O Box 210-40601, Bondo, Kenya
| |
Collapse
|
30
|
Chia S, Xia J, Kwan YH, Lim ZY, Tan CS, Low SG, Xu B, Loo YX, Kong LY, Koh CW, Towle RM, Lim SF, Yoon S, Seah SSY, Low LL. Evaluating the association of COVID-19 restrictions on discharge planning and post-discharge outcomes in the community hospital and Singapore regional health system. FRONTIERS IN HEALTH SERVICES 2023; 3:1147698. [PMID: 37744642 PMCID: PMC10513784 DOI: 10.3389/frhs.2023.1147698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/24/2023] [Indexed: 09/26/2023]
Abstract
Objectives The COVID-19 is a global health issue with widespread impact around the world, and many countries initiated lockdowns as part of their preventive measures. We aim to quantify the duration of delay in discharge to community from Community Hospitals, as well as quantify adverse patient outcomes post discharge pre and during lockdown period. Design and methods We conducted a before-after study comparing the length of stay in Community Hospitals, unscheduled readmissions or Emergency Department attendance, patients' quality of life using EQ5D-5l, number and severity of falls, in patients admitted and discharged before and during lockdown period. Results The average length of stay in the lockdown group (27.77 days) were significantly longer than that of the pre-lockdown group (23.76 days), p = 0.003. There were similar proportions of patients with self-reported falls post discharge between both groups. Patients in the pre-lockdown group had slightly better EQ-5D-5l Index score at 0.55, compared to the lockdown study group at 0.49. Half of the patients in both groups were referred to Community Care Services on discharge. Conclusion Our study would help in developing a future systematic preparedness guideline and contingency plans in times of disease outbreak and other similar public health emergencies.
Collapse
Affiliation(s)
- Shermain Chia
- Post-Acute and Continuing Care, Outram Community Hospital, Singapore, Singapore
| | - Jiawen Xia
- Research and Translational Innovation Office, Singhealth Community Hospitals, Singapore, Singapore
| | - Yu Heng Kwan
- Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Zhui Ying Lim
- Population Health & Integrated Care Office, Singapore General Hospital, Singapore, Singapore
| | - Chuen Seng Tan
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Sher Guan Low
- Post-Acute and Continuing Care, Sengkang Community Hospital, Singapore, Singapore
| | - Bangyu Xu
- Post-Acute and Continuing Care, Sengkang Community Hospital, Singapore, Singapore
| | - Yu Xian Loo
- Post-Acute and Continuing Care, Outram Community Hospital, Singapore, Singapore
| | - Lai Yee Kong
- Medical Social Services, Sengkang Community Hospital, Singapore, Singapore
| | - Chee Wai Koh
- Medical Social Services, Outram Community Hospital, Singapore, Singapore
| | - Rachel Marie Towle
- Specialty Nursing, Population Health & Integrated Care Office, Regional Health System, Singapore, Singapore
| | - Su Fee Lim
- Regional Health System Community Nursing, Population Health & Integrated Care Office, Singapore General Hospital, Singapore, Singapore
| | - Sungwon Yoon
- Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
| | - Sharna Si Ying Seah
- Research and Translational Innovation Office, Singhealth Community Hospitals, Singapore, Singapore
| | - Lian Leng Low
- Post-Acute and Continuing Care, Outram Community Hospital, Singapore, Singapore
| |
Collapse
|
31
|
Williamson SF, Williams CJ, Lendrem BC, Wilson KJ. Sample size determination for point-of-care COVID-19 diagnostic tests: a Bayesian approach. Diagn Progn Res 2023; 7:17. [PMID: 37596684 PMCID: PMC10436636 DOI: 10.1186/s41512-023-00153-1] [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: 01/10/2023] [Accepted: 07/14/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND In a pandemic setting, it is critical to evaluate and deploy accurate diagnostic tests rapidly. This relies heavily on the sample size chosen to assess the test accuracy (e.g. sensitivity and specificity) during the diagnostic accuracy study. Too small a sample size will lead to imprecise estimates of the accuracy measures, whereas too large a sample size may delay the development process unnecessarily. This study considers use of a Bayesian method to guide sample size determination for diagnostic accuracy studies, with application to COVID-19 rapid viral detection tests. Specifically, we investigate whether utilising existing information (e.g. from preceding laboratory studies) within a Bayesian framework can reduce the required sample size, whilst maintaining test accuracy to the desired precision. METHODS The method presented is based on the Bayesian concept of assurance which, in this context, represents the unconditional probability that a diagnostic accuracy study yields sensitivity and/or specificity intervals with the desired precision. We conduct a simulation study to evaluate the performance of this approach in a variety of COVID-19 settings, and compare it to commonly used power-based methods. An accompanying interactive web application is available, which can be used by researchers to perform the sample size calculations. RESULTS Results show that the Bayesian assurance method can reduce the required sample size for COVID-19 diagnostic accuracy studies, compared to standard methods, by making better use of laboratory data, without loss of performance. Increasing the size of the laboratory study can further reduce the required sample size in the diagnostic accuracy study. CONCLUSIONS The method considered in this paper is an important advancement for increasing the efficiency of the evidence development pathway. It has highlighted that the trade-off between lab study sample size and diagnostic accuracy study sample size should be carefully considered, since establishing an adequate lab sample size can bring longer-term gains. Although emphasis is on its use in the COVID-19 pandemic setting, where we envisage it will have the most impact, it can be usefully applied in other clinical areas.
Collapse
Affiliation(s)
- S Faye Williamson
- Biostatistics Research Group, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.
| | - Cameron J Williams
- NIHR Newcastle In Vitro Diagnostic Cooperative, Newcastle University, Newcastle upon Tyne, UK
| | - B Clare Lendrem
- NIHR Newcastle In Vitro Diagnostic Cooperative, Newcastle University, Newcastle upon Tyne, UK
| | - Kevin J Wilson
- School of Mathematics, Statistics and Physics, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
32
|
Seaman WT, Keener O, Mei W, Mollan KR, Jones CD, Pettifor A, Bowman NM, Wang F, Webster-Cyriaque J. Oral SARS-CoV-2 host responses predict the early COVID-19 disease course. RESEARCH SQUARE 2023:rs.3.rs-3154698. [PMID: 37645853 PMCID: PMC10462189 DOI: 10.21203/rs.3.rs-3154698/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Objectives Oral fluids provide ready detection of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses. This study sought to determine relationships between oral virus, oral anti-SARS-CoV-2-specific antibodies, and symptoms. Methods Saliva/throat wash (saliva/TW) were collected from asymptomatic and symptomatic, nasopharyngeal (NP) SARS-CoV-2 RT-qPCR+, subjects (n=47). SARS-CoV-2 RT-qPCR, N-antigen detection by immunoblot and lateral flow assay (LFA) were performed. RT-qPCR targeting viral subgenomic RNA (sgRNA) was sequence confirmed. SARS-CoV-2-anti-S protein RBD LFA assessed IgM and IgG responses. Structural analysis identified host salivary molecules analogous to SARS-CoV-2-N-antigen. Statistical analyses were performed. Results At baseline, LFA-detected N-antigen was immunoblot-confirmed in 82% of TW. However, only 3/17 were saliva/TW qPCR+. Sixty percent of saliva and 83% of TW demonstrated persistent N-antigen at 4 weeks. N-antigen LFA signal in three negative subjects suggested potential cross-detection of 4 structurally analogous salivary RNA binding proteins (alignment 19-29aa, RMSD 1-1.5 Angstroms). At entry, symptomatic subjects demonstrated replication-associated sgRNA junctions, were IgG+ (94%/100% in saliva/TW), and IgM+ (75%/63%). At 4 weeks, SARS-CoV-2 IgG (100%/83%) and IgM (80%/67%) persisted. Oral IgG correlated 100% with NP+PCR status. Cough and fatigue severity (p=0.0008 and 0.016), and presence of nausea, weakness, and composite upper respiratory symptoms (p=0.005, 0.037 and 0.017) were negatively associated with oral IgM. Female oral IgM levels were higher than male (p=0.056). Conclusion Important to transmission and disease course, oral viral replication and persistence showed clear relationships with select symptoms, early Ig responses, and gender during early infection. N-antigen cross-reactivity may reflect mimicry of structurally analogous host proteins.
Collapse
Affiliation(s)
- William T Seaman
- National Institute of Dental and Craniofacial Research, National Institutes of Health
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Vierbaum L, Wojtalewicz N, Grunert HP, Zimmermann A, Scholz A, Goseberg S, Kaiser P, Duehring U, Drosten C, Corman V, Niemeyer D, Rabenau HF, Obermeier M, Nitsche A, Michel J, Puyskens A, Huggett JF, O'Sullivan DM, Busby E, Cowen S, Vallone PM, Cleveland MH, Falak S, Kummrow A, Schellenberg I, Zeichhardt H, Kammel M. Results of German external quality assessment schemes for SARS-CoV-2 antigen detection. Sci Rep 2023; 13:13206. [PMID: 37580353 PMCID: PMC10425338 DOI: 10.1038/s41598-023-40330-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 08/08/2023] [Indexed: 08/16/2023] Open
Abstract
The COVID-19 pandemic illustrated the important role of diagnostic tests, including lateral flow tests (LFTs), in identifying patients and their contacts to slow the spread of infections. INSTAND performed external quality assessments (EQA) for SARS-CoV-2 antigen detection with lyophilized and chemically inactivated cell culture supernatant of SARS-CoV-2 infected Vero cells. A pre-study demonstrated the suitability of the material. Participants reported qualitative and/or quantitative antigen results using either LFTs or automated immunoassays for five EQA samples per survey. 711 data sets were reported for LFT detection in three surveys in 2021. This evaluation focused on the analytical sensitivity of different LFTs and automated immunoassays. The inter-laboratory results showed at least 94% correct results for non-variant of concern (VOC) SARS-CoV-2 antigen detection for viral loads of ≥ 4.75 × 106 copies/mL and SARS-CoV-2 negative samples. Up to 85% had success for a non-VOC viral load of ~ 1.60 × 106 copies/mL. A viral load of ~ 1.42 × 107 copies/mL of the Delta VOC was reported positive in > 96% of results. A high specificity was found with almost 100% negative SARS-CoV-2 antigen results for HCoV 229E and HCoV NL63 positive samples. Quantitative results correlated with increasing SARS-CoV-2 viral load but showed a broad scatter. This study shows promising SARS-CoV-2 antigen test performance of the participating laboratories, but further investigations with the now predominant Omicron VOC are needed.
Collapse
Affiliation(s)
- Laura Vierbaum
- INSTAND E.V., Society for Promoting Quality Assurance in Medical Laboratories, Ubierstr. 20, 40223, Düsseldorf, Germany.
| | - Nathalie Wojtalewicz
- INSTAND E.V., Society for Promoting Quality Assurance in Medical Laboratories, Ubierstr. 20, 40223, Düsseldorf, Germany
| | - Hans-Peter Grunert
- GBD Gesellschaft für Biotechnologische Diagnostik mbH, Berlin, Potsdamer Chaussee 80, 14129, Berlin, Germany
| | - Anika Zimmermann
- IQVD GmbH, Institut für Qualitätssicherung in der Virusdiagnostik, Berlin, Potsdamer Chaussee 80, 14129, Berlin, Germany
| | - Annemarie Scholz
- IQVD GmbH, Institut für Qualitätssicherung in der Virusdiagnostik, Berlin, Potsdamer Chaussee 80, 14129, Berlin, Germany
| | - Sabine Goseberg
- INSTAND E.V., Society for Promoting Quality Assurance in Medical Laboratories, Ubierstr. 20, 40223, Düsseldorf, Germany
| | - Patricia Kaiser
- INSTAND E.V., Society for Promoting Quality Assurance in Medical Laboratories, Ubierstr. 20, 40223, Düsseldorf, Germany
| | - Ulf Duehring
- GBD Gesellschaft für Biotechnologische Diagnostik mbH, Berlin, Potsdamer Chaussee 80, 14129, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Charité - University Medicine Berlin; National Consultant Laboratory for Coronaviruses; German Centre for Infection Research, Berlin, Berlin, Germany
| | - Victor Corman
- Institute of Virology, Charité - University Medicine Berlin; National Consultant Laboratory for Coronaviruses; German Centre for Infection Research, Berlin, Berlin, Germany
| | - Daniela Niemeyer
- Institute of Virology, Charité - University Medicine Berlin; National Consultant Laboratory for Coronaviruses; German Centre for Infection Research, Berlin, Berlin, Germany
| | - Holger F Rabenau
- Institute of Virology, Charité - University Medicine Berlin; National Consultant Laboratory for Coronaviruses; German Centre for Infection Research, Berlin, Berlin, Germany
| | - Martin Obermeier
- Institute for Medical Virology, University Hospital, Goethe University Frankfurt, Frankfurt, Hessen, Germany
| | | | - Janine Michel
- Robert Koch Institute, Highly Pathogenic Viruses, Centre for Biological Threats and Special Pathogens, WHO Reference Laboratory for SARS-CoV-2 and WHO Collaborating Centre for Emerging Infections and Biological Threats, Robert Koch Institute, Berlin, Germany
| | - Andreas Puyskens
- Robert Koch Institute, Highly Pathogenic Viruses, Centre for Biological Threats and Special Pathogens, WHO Reference Laboratory for SARS-CoV-2 and WHO Collaborating Centre for Emerging Infections and Biological Threats, Robert Koch Institute, Berlin, Germany
| | - Jim F Huggett
- National Measurement Laboratory, LGC, Teddington, Middlesex, UK
- School of Biosciences and Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK
| | | | - Eloise Busby
- National Measurement Laboratory, LGC, Teddington, Middlesex, UK
| | - Simon Cowen
- National Measurement Laboratory, LGC, Teddington, Middlesex, UK
| | - Peter M Vallone
- NIST, National Institute of Standards and Technology, Applied Genetics Group, Biomolecular Measurement Division, Materials Measurement Laboratory, Gaithersburg, MD, USA
| | - Megan H Cleveland
- NIST, National Institute of Standards and Technology, Applied Genetics Group, Biomolecular Measurement Division, Materials Measurement Laboratory, Gaithersburg, MD, USA
| | - Samreen Falak
- Physikalisch-Technische Bundesanstalt, Berlin, Germany
| | | | - Ingo Schellenberg
- INSTAND E.V., Society for Promoting Quality Assurance in Medical Laboratories, Ubierstr. 20, 40223, Düsseldorf, Germany
- Institute of Bioanalytical Sciences, Center of Life Sciences, Anhalt University of Applied Sciences, Bernburg, Saxony-Anhalt, Germany
| | - Heinz Zeichhardt
- INSTAND E.V., Society for Promoting Quality Assurance in Medical Laboratories, Ubierstr. 20, 40223, Düsseldorf, Germany
- IQVD GmbH, Institut für Qualitätssicherung in der Virusdiagnostik, Berlin, Potsdamer Chaussee 80, 14129, Berlin, Germany
- GBD Gesellschaft für Biotechnologische Diagnostik mbH, Berlin, Potsdamer Chaussee 80, 14129, Berlin, Germany
| | - Martin Kammel
- INSTAND E.V., Society for Promoting Quality Assurance in Medical Laboratories, Ubierstr. 20, 40223, Düsseldorf, Germany
- IQVD GmbH, Institut für Qualitätssicherung in der Virusdiagnostik, Berlin, Potsdamer Chaussee 80, 14129, Berlin, Germany
| |
Collapse
|
34
|
Seaman WT, Keener O, Mei W, Mollan KR, Jones CD, Pettifor A, Bowman NM, Wang F, Webster-Cyriaque J. Oral SARS-CoV-2 host responses predict the early COVID-19 disease course. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.06.23286853. [PMID: 37609199 PMCID: PMC10441495 DOI: 10.1101/2023.03.06.23286853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Objectives Oral fluids provide ready detection of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses. This study sought to determine relationships between oral virus, oral anti-SARS-CoV-2-specific antibodies, and symptoms. Methods Saliva/throat wash (saliva/TW) were collected from asymptomatic and symptomatic, nasopharyngeal (NP) SARS-CoV-2 RT-qPCR+, subjects (n=47). SARS-CoV-2 RT-qPCR, N-antigen detection by immunoblot and lateral flow assay (LFA) were performed. RT-qPCR targeting viral subgenomic RNA (sgRNA) was sequence confirmed. SARS-CoV-2-anti-S protein RBD LFA assessed IgM and IgG responses. Structural analysis identified host salivary molecules analogous to SARS-CoV-2-N-antigen. Statistical analyses were performed. Results At baseline, LFA-detected N-antigen was immunoblot-confirmed in 82% of TW. However, only 3/17 were saliva/TW qPCR+. Sixty percent of saliva and 83% of TW demonstrated persistent N-antigen at 4 weeks. N-antigen LFA signal in three negative subjects suggested potential cross-detection of 4 structurally analogous salivary RNA binding proteins (alignment 19-29aa, RMSD 1-1.5 Angstroms). At entry, symptomatic subjects demonstrated replication-associated sgRNA junctions, were IgG+ (94%/100% in saliva/TW), and IgM+ (75%/63%). At 4 weeks, SARS-CoV-2 IgG (100%/83%) and IgM (80%/67%) persisted. Oral IgG correlated 100% with NP+PCR status. Cough and fatigue severity (p=0.0008 and 0.016), and presence of nausea, weakness, and composite upper respiratory symptoms (p=0.005, 0.037 and 0.017) were negatively associated with oral IgM. Female oral IgM levels were higher than male (p=0.056). Conclusion Important to transmission and disease course, oral viral replication and persistence showed clear relationships with select symptoms, early Ig responses, and gender during early infection. N-antigen cross-reactivity may reflect mimicry of structurally analogous host proteins.
Collapse
|
35
|
Yang YP, Jiesisibieke ZL, Tung TH. Association Between Rapid Antigen Detection Tests and Real-Time Reverse Transcription-Polymerase Chain Reaction Assay for SARS-CoV-2: A Systematic Review and Meta-Analyses. Int J Public Health 2023; 68:1605452. [PMID: 37588042 PMCID: PMC10425602 DOI: 10.3389/ijph.2023.1605452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 07/11/2023] [Indexed: 08/18/2023] Open
Abstract
Objectives: We aimed to assess the association between rapid antigen detection tests and real-time reverse transcription-polymerase chain reaction assay for severe acute respiratory syndrome coronavirus 2. Methods: We searched PubMed, Cochrane Library, EMBASE, and the Web of Science from their inception to 31 May 2023. A random-effects meta-analysis was used to estimate false positives in the RADTs group, relative to those in the RT-PCR group, and subgroup analyses were conducted based on the different Ct value cut-offs (<40 or ≥40). We performed this study in accordance with the guidelines outlined in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). Results: Fifty-one studies were included and considered to be of moderate quality. We found a satisfactory overall false positive rate (0.01, 95% CI: 0.00-0.01) for the RADTs compared to RT-PCR. In the stratified analysis, we also found that the false positive rates of the RADTs did not increase when Ct values of RT-PCR (Ct < 40, 0.01, 95% CI: 0.00-0.01; Ct ≥ 40, 0.01, 95% CI: 0.00-0.01). Conclusion: In conclusion, the best available evidence supports an association between RADTs and RT-PCR. When Ct-values were analyzed using cut-off <40 or ≥40, this resulted in an estimated false positive rate of only 1%.
Collapse
Affiliation(s)
- Yu-Pei Yang
- Department of Hematology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Zhu Liduzi Jiesisibieke
- School of Public Health, The University of Hong Kong Li Ka Shing Faculty of Medicine, Pokfulam, Hong Kong, Hong Kong SAR, China
| | - Tao-Hsin Tung
- Evidence-Based Medicine Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| |
Collapse
|
36
|
Samsunder N, Lustig G, Ngubane S, Maseko TG, Rambaran S, Ngcapu S, Magini SN, Lewis L, Cawood C, Kharsany ABM, Karim QA, Karim SA, Naidoo K, Sivro A. Field evaluations of four SARS-CoV-2 rapid antigen tests during SARS-CoV-2 Delta variant wave in South Africa. Diagn Progn Res 2023; 7:14. [PMID: 37491317 PMCID: PMC10369830 DOI: 10.1186/s41512-023-00151-3] [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: 12/12/2022] [Accepted: 07/11/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Rapid antigen tests detecting SARS-CoV-2 were shown to be a useful tool in managing the COVID-19 pandemic. Here, we report on the results of a prospective diagnostic accuracy study of four SARS-CoV-2 rapid antigen tests in a South African setting. METHODS Rapid antigen test evaluations were performed through drive-through testing centres in Durban, South Africa, from July to December 2021. Two evaluation studies were performed: nasal Panbio COVID-19 Ag Rapid Test Device (Abbott) was evaluated in parallel with the nasopharyngeal Espline SARS-CoV-2 Ag test (Fujirebio), followed by the evaluation of nasal RightSign COVID-19 Antigen Rapid test Cassette (Hangzhou Biotest Biotech) in parallel with the nasopharyngeal STANDARD Q COVID-19 Ag test (SD Biosensor). The Abbott RealTime SARS-CoV-2 assay was used as a reference test. RESULTS Evaluation of Panbio and Espline Ag tests was performed on 494 samples (31% positivity), while the evaluation of Standard Q and RightTest Ag tests was performed on 539 samples (13.17% positivity). The overall sensitivity for all four tests ranged between 60 and 72% with excellent specificity values (> 98%). Sensitivity increased to > 80% in all tests in samples with cycle number value < 20. All four tests performed best in samples from patients presenting within the first week of symptom onset. CONCLUSIONS All four evaluated tests detected a majority of the cases within the first week of symptom onset with high viral load.
Collapse
Affiliation(s)
- Natasha Samsunder
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Gila Lustig
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Slindile Ngubane
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Thando Glory Maseko
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Santhuri Rambaran
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Sinaye Ngcapu
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| | - Stanley Nzuzo Magini
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Lara Lewis
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Cherie Cawood
- Epicentre AIDS Risk Management (Pty) Limited, Durban, South Africa
| | - Ayesha B M Kharsany
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| | - Quarraisha Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Epidemiology, Columbia University, New York City, USA
| | - Salim Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Epidemiology, Columbia University, New York City, USA
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- South African Medical Research Council (SAMRC), CAPRISA-TB-HIV Pathogenesis and Treatment Research Unit, University of KwaZulu-Natal Nelson R. Mandela School of Medicine, Durban, South Africa
| | - Aida Sivro
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa.
- South African Medical Research Council (SAMRC), CAPRISA-TB-HIV Pathogenesis and Treatment Research Unit, University of KwaZulu-Natal Nelson R. Mandela School of Medicine, 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.
| |
Collapse
|
37
|
Golden A, Cantera JL, Lillis L, Phan TT, Slater H, Webb EJ, Peck RB, Boyle DS, Domingo GJ. A Reagent and Virus Benchmarking Panel for a Uniform Analytical Performance Assessment of N Antigen-Based Diagnostic Tests for COVID-19. Microbiol Spectr 2023; 11:e0373122. [PMID: 37166329 PMCID: PMC10269465 DOI: 10.1128/spectrum.03731-22] [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: 09/28/2022] [Accepted: 04/21/2023] [Indexed: 05/12/2023] Open
Abstract
Rapid diagnostic tests (RDTs) that detect antigen indicative of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection can help in making quick health care decisions and regularly monitoring groups at risk of infection. With many RDT products entering the market, it is important to rapidly evaluate their relative performance. Comparison of clinical evaluation study results is challenged by protocol design variations and study populations. Laboratory assays were developed to quantify nucleocapsid (N) and spike (S) SARS-CoV-2 antigens. Quantification of the two antigens in nasal eluates confirmed higher abundance of N than S antigen. The median concentration of N antigen was 10 times greater than S per genome equivalent. The N antigen assay was used in combination with quantitative reverse transcription (RT)-PCR to qualify a panel composed of recombinant antigens, inactivated virus, and clinical specimen pools. This benchmarking panel was applied to evaluate the analytical performance of the SD Biosensor Standard Q COVID-19 antigen (Ag) test, Abbott Panbio COVID-19 Ag rapid test, Abbott BinaxNOW COVID-19 Ag test, and the LumiraDx SARS-CoV-2 Ag test. The four tests displayed different sensitivities toward the different panel members, but all performed best with the clinical specimen pool. The concentration for a 90% probability of detection across the four tests ranged from 21 to 102 pg/mL of N antigen in the extracted sample. Benchmarking panels provide a quick way to verify the baseline performance of a diagnostic and enable direct comparisons between diagnostic tests. IMPORTANCE This study reports the results for severe acute respiratory syndrome coronavirus-2 (SARS-COV-2) nucleocapsid (N) and spike (S) antigen quantification assays and their performance against clinical reverse transcription (RT)-PCR results, thus describing an open-access quantification method for two important SARS-CoV-2 protein analytes. Characterized N antigen panels were used to evaluate the limits of detection of four different rapid tests for SARS-CoV-2 against multiple sources of nucleocapsid antigen, demonstrating proof-of-concept materials and methodology to evaluate SARS-CoV-2 rapid antigen detection tests. Quantification of N antigen was used to characterize the relationship between viral count and antigen concentration among clinical samples and panel members of both clinical sample and viral culture origin. This contributes to a deeper understanding of protein antigen and molecular analytes and presents analytical methods complementary to clinical evaluation for characterizing the performance of both laboratory-based and point-of-care rapid diagnostics for SARS-CoV-2.
Collapse
|
38
|
Dong T, Wang M, Liu J, Ma P, Pang S, Liu W, Liu A. Diagnostics and analysis of SARS-CoV-2: current status, recent advances, challenges and perspectives. Chem Sci 2023; 14:6149-6206. [PMID: 37325147 PMCID: PMC10266450 DOI: 10.1039/d2sc06665c] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 05/03/2023] [Indexed: 06/17/2023] Open
Abstract
The disastrous spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has induced severe public healthcare issues and weakened the global economy significantly. Although SARS-CoV-2 infection is not as fatal as the initial outbreak, many infected victims suffer from long COVID. Therefore, rapid and large-scale testing is critical in managing patients and alleviating its transmission. Herein, we review the recent advances in techniques to detect SARS-CoV-2. The sensing principles are detailed together with their application domains and analytical performances. In addition, the advantages and limits of each method are discussed and analyzed. Besides molecular diagnostics and antigen and antibody tests, we also review neutralizing antibodies and emerging SARS-CoV-2 variants. Further, the characteristics of the mutational locations in the different variants with epidemiological features are summarized. Finally, the challenges and possible strategies are prospected to develop new assays to meet different diagnostic needs. Thus, this comprehensive and systematic review of SARS-CoV-2 detection technologies may provide insightful guidance and direction for developing tools for the diagnosis and analysis of SARS-CoV-2 to support public healthcare and effective long-term pandemic management and control.
Collapse
Affiliation(s)
- Tao Dong
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University 308 Ningxia Road Qingdao 266071 China
- School of Pharmacy, Medical College, Qingdao University 308 Ningxia Road Qingdao 266071 China
| | - Mingyang Wang
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University 308 Ningxia Road Qingdao 266071 China
| | - Junchong Liu
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University 308 Ningxia Road Qingdao 266071 China
| | - Pengxin Ma
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University 308 Ningxia Road Qingdao 266071 China
| | - Shuang Pang
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University 308 Ningxia Road Qingdao 266071 China
| | - Wanjian Liu
- Qingdao Hightop Biotech Co., Ltd 369 Hedong Road, Hi-tech Industrial Development Zone Qingdao 266112 China
| | - Aihua Liu
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University 308 Ningxia Road Qingdao 266071 China
| |
Collapse
|
39
|
Abstract
Nucleic acid amplification tests (NAATs), including polymerase chain reaction (PCR) assays, are more sensitive for the detection of SARS-CoV-2 than rapid antigen tests (RATS), and are the gold standard for diagnosis of acute COVID-19. However NAATs can remain positive for weeks following infection due to low-level shedding of non-viable viral fragments. RATs (in particular self-testing) are the mainstay of COVID-19 diagnosis due to their convenience, speed and high specificity. The sensitivity of RATs is highest within seven days of symptom onset. A negative RAT result may warrant a NAAT or repeat RAT for confirmation. The presence of spike antibodies is consistent with either vaccination or infection. Nucleocapsid antibodies suggest a previous infection. Serological tests measuring neutralising antibodies that infer immunity are not readily available.
Collapse
|
40
|
Nasrallah GK, Ali F, Younes S, Al Khatib HA, Al-Thani AA, Yassine HM. Enhancing the sensitivity of rapid antigen detection test (RADT) of different SARS-CoV-2 variants and lineages using fluorescence-labeled antibodies and a fluorescent meter. Heliyon 2023; 9:e17179. [PMID: 37325455 PMCID: PMC10257515 DOI: 10.1016/j.heliyon.2023.e17179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/21/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023] Open
Abstract
RT-qPCR is considered the gold standard for diagnosis of COVID-19; however, it is laborious, time-consuming, and expensive. RADTs have evolved recently as relatively inexpensive methods to address these shortcomings, but their performance for detecting different SARS-COV-2 variants remains limited. RADT test performance could be enhanced using different antibody labeling and signal detection techniques. Here, we aimed to evaluate the performance of two antigen RADTs for detecting different SARS-CoV-2 variants: (i) the conventional colorimetric RADT (Ab-conjugated with gold beads); and (ii) the new Finecare™ RADT (Ab-coated fluorescent beads). Finecare™ is a meter used for the detection of a fluorescent signal. 187 frozen nasopharyngeal swabs collected in Universal transport (UTM) that are RT-qPCR positive for different SARS-CoV-2 variants were selected, including Alpha (n = 60), Delta (n = 59), and Omicron variants (n = 108). Sixty flu and 60 RSV-positive samples were included as negative controls (total sample number = 347). The conventional RADT showed sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) of 62.4% (95%CI: 54-70), 100% (95%CI: 97-100), 100% (95%CI: 100-100), and 58% (95%CI: 49-67), respectively. These measurements were enhanced using the Finecare™ RADT: sensitivity, specificity, PPV, and NPV were 92.6% (95%CI: 89.08-92.3), 96% (95%CI: 96-99.61), 98% (95%CI: 89-92.3), and 85% (95%CI: 96-99.6) respectively. The sensitivity of both RADTs could be greatly underestimated because nasopharyngeal swab samples collected UTM and stored at -80 °C were used. Despite that, our results indicate that the Finecare™ RADT is appropriate for clinical laboratory and community-based surveillance due to its high sensitivity and specificity.
Collapse
Affiliation(s)
- Gheyath K Nasrallah
- Biomedical Research Center, Qatar University, Doha, 2713, Qatar
- Biomedical Sciences Department, College of Health Sciences, Qatar University, Doha, 2713, Qatar
| | - Fatma Ali
- Biomedical Research Center, Qatar University, Doha, 2713, Qatar
- Biomedical Sciences Department, College of Health Sciences, Qatar University, Doha, 2713, Qatar
| | - Salma Younes
- Biomedical Research Center, Qatar University, Doha, 2713, Qatar
- Biomedical Sciences Department, College of Health Sciences, Qatar University, Doha, 2713, Qatar
| | | | | | - Hadi M Yassine
- Biomedical Research Center, Qatar University, Doha, 2713, Qatar
- Biomedical Sciences Department, College of Health Sciences, Qatar University, Doha, 2713, Qatar
| |
Collapse
|
41
|
Bos DAG, Lagrou K, Verbakel JY. Prospective Performance Evaluation of the miDiagnostics COVID-19 PCR Test for Rapid SARS-CoV-2 Detection on Nasopharyngeal Swabs. J Clin Microbiol 2023; 61:e0187122. [PMID: 37093001 PMCID: PMC10204629 DOI: 10.1128/jcm.01871-22] [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: 12/22/2022] [Accepted: 03/27/2023] [Indexed: 04/25/2023] Open
Abstract
Rapid diagnosis or exclusion of SARS-CoV-2 infection is essential for correct medical management decisions regarding COVID-19. High-throughput laboratory-based reverse transcriptase (RT)-PCR testing is accurate with longer turnaround times, while rapid antigen tests show moderate sensitivity. In search of a fast and reliable COVID-19 test, we aimed to validate the rapid miDiagnostics COVID-19 PCR test. We recruited symptomatic and asymptomatic participants in a mobile COVID-19 test center in Belgium. We collected three nasopharyngeal samples from each participant. The index sample was tested on the miDiagnostics COVID-19 PCR reader, the reference sample was tested on the reference TaqPath COVID-19 PCR test in the Belgian Reference Center for Respiratory Pathogens of University Hospitals Leuven, and a third sample was collected for discordance testing with the PerkinElmer SARS-CoV-2 PCR kit. A total of 770 participants yielded 763 sets of included nasopharyngeal samples. Overall positive percent agreement and negative percent agreement of the miDiagnostics COVID-19 PCR test were 95.5% (92.6% to 97.4%) and 94.9% (92.3 to 96.8%), rising to 98.6% (96.5% to 99.6%) and 96.5% (92.6% to 98.7%) in symptomatic patients. Discordance testing reclassified 15 of 21 false-positive cases as true positive. A retest of the miDiagnostics PCR test was performed in 61 tests (7.4%) due to a technical error. The miDiagnostics COVID-19 PCR test showed excellent clinical accuracy. The fast and reliable results allow for rapid correct diagnosis and tailored medical management decisions regarding COVID-19.
Collapse
Affiliation(s)
- David A. G. Bos
- EPI-Centre, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Katrien Lagrou
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jan Y. Verbakel
- EPI-Centre, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
- NIHR Community Healthcare Medtech and IVD cooperative, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
42
|
Newbold SC, Ashworth M, Finnoff D, Shogren JF, Thunström L. Physical distancing versus testing with self-isolation for controlling an emerging epidemic. Sci Rep 2023; 13:8185. [PMID: 37210388 DOI: 10.1038/s41598-023-35083-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 05/12/2023] [Indexed: 05/22/2023] Open
Abstract
Two distinct strategies for controlling an emerging epidemic are physical distancing and regular testing with self-isolation. These strategies are especially important before effective vaccines or treatments become widely available. The testing strategy has been promoted frequently but used less often than physical distancing to mitigate COVID-19. We compared the performance of these strategies in an integrated epidemiological and economic model that includes a simple representation of transmission by "superspreading," wherein a relatively small fraction of infected individuals cause a large share of infections. We examined the economic benefits of distancing and testing over a wide range of conditions, including variations in the transmissibility and lethality of the disease meant to encompass the most prominent variants of COVID-19 encountered so far. In a head-to-head comparison using our primary parameter values, both with and without superspreading and a declining marginal value of mortality risk reductions, an optimized testing strategy outperformed an optimized distancing strategy. In a Monte Carlo uncertainty analysis, an optimized policy that combined the two strategies performed better than either one alone in more than 25% of random parameter draws. Insofar as diagnostic tests are sensitive to viral loads, and individuals with high viral loads are more likely to contribute to superspreading events, superspreading enhances the relative performance of testing over distancing in our model. Both strategies performed best at moderate levels of transmissibility, somewhat lower than the transmissibility of the ancestral strain of SARS-CoV-2.
Collapse
Affiliation(s)
- Stephen C Newbold
- Department of Economics, University of Wyoming, Laramie, WY, 82071, USA.
| | - Madison Ashworth
- Department of Economics, University of Wyoming, Laramie, WY, 82071, USA
- Fletcher Group, Inc., London, KY, 40741, USA
| | - David Finnoff
- Department of Economics, University of Wyoming, Laramie, WY, 82071, USA
| | - Jason F Shogren
- Department of Economics, University of Wyoming, Laramie, WY, 82071, USA
| | - Linda Thunström
- Department of Economics, University of Wyoming, Laramie, WY, 82071, USA
| |
Collapse
|
43
|
Jubair M, Rahman S, Begum MN, Talha M, Musarrat R, Rahman AKMS, Uddin MS, Wen A, Ning Y, Nahar K, Rahman MZ, Rahman M. SARS-CoV-2 antigen detection by saliva; an alternative to nasopharyngeal specimen: A cross-sectional study. Health Sci Rep 2023; 6:e1275. [PMID: 37216057 PMCID: PMC10196438 DOI: 10.1002/hsr2.1275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
Background and Aims Saliva samples are less invasive and more convenient for patients than naso- and/or oropharynx swabs (NOS). However, there is no US Food and Drug Administration-approved severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) rapid antigen test kit, which can be useful in a prolonged pandemic to reduce transmission by allowing suspected individuals to self-sampling. We evaluated the performances of High sensitive AQ+ Rapid SARS-CoV-2 Antigen Test (AQ+ kit) using nasopharyngeal swabs (NPs) and saliva specimens from the same patients in laboratory conditions. Methods The real-time reverse transcription-polymerase chain reaction (rRT-PCR) test result was used for screening the inrolled individuals and compared as the gold standard. NP and saliva samples were collected from 100 rRT-PCR positives and 100 negative individuals and tested with an AQ+ kit. Results The AQ+ kit showed good performances in both NP and saliva samples with an overall accuracy of 98.5% and 94.0%, and sensitivity of 97.0% and 88.0%, respectively. In both cases, specificity was 100%. AQ+ kit performance with saliva was in the range of the World Health Organization recommended value. Conclusion xOur findings indicate that the saliva specimen can be used as an alternative and less invasive to NPs for quick and reliable SARS-CoV-2 antigen detection.
Collapse
Affiliation(s)
- Mohammad Jubair
- Virology Laboratory, Infectious Diseases Divisionicddr,b: International Centre for Diarrhoeal Disease Research, BangladeshDhakaBangladesh
| | - Sezanur Rahman
- Virology Laboratory, Infectious Diseases Divisionicddr,b: International Centre for Diarrhoeal Disease Research, BangladeshDhakaBangladesh
| | - Mst. Noorjahan Begum
- Virology Laboratory, Infectious Diseases Divisionicddr,b: International Centre for Diarrhoeal Disease Research, BangladeshDhakaBangladesh
| | - Muhammad Talha
- Virology Laboratory, Infectious Diseases Divisionicddr,b: International Centre for Diarrhoeal Disease Research, BangladeshDhakaBangladesh
| | - Raisha Musarrat
- Virology Laboratory, Infectious Diseases Divisionicddr,b: International Centre for Diarrhoeal Disease Research, BangladeshDhakaBangladesh
| | | | | | | | | | - Kamrun Nahar
- Virology Laboratory, Infectious Diseases Divisionicddr,b: International Centre for Diarrhoeal Disease Research, BangladeshDhakaBangladesh
| | - Mohammed Ziaur Rahman
- Virology Laboratory, Infectious Diseases Divisionicddr,b: International Centre for Diarrhoeal Disease Research, BangladeshDhakaBangladesh
| | - Mustafizur Rahman
- Virology Laboratory, Infectious Diseases Divisionicddr,b: International Centre for Diarrhoeal Disease Research, BangladeshDhakaBangladesh
| |
Collapse
|
44
|
Campbell WS, Donahue M, Williams RM, McCutchen E, Broadhurst J, Schnaubelt A, Staffend NJ, Hinrichs SH, Iwen PC. A Public Health Laboratory Information System in Support of Health Emergencies: The Nebraska Public Health Laboratory COVID-19 Experience. Public Health Rep 2023:333549231168459. [PMID: 37125740 PMCID: PMC10133861 DOI: 10.1177/00333549231168459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
OBJECTIVES Public health laboratories (PHLs) are essential components of US Public Health Service operations. The health information technology that supports PHLs is central to effective and efficient laboratory operations and overall public health response to infectious disease management. This analysis presents key information on how the Nebraska Public Health Laboratory (NPHL) information technology system evolved to meet the demands of the COVID-19 pandemic. MATERIALS AND METHODS COVID-19 presented numerous, unforeseen information technology system challenges. The most notable challenges requiring changes to NPHL software systems and capability were improving efficiency of the laboratory operation due to high-volume testing, responding daily to demands for timely data for analysis by partner systems, interfacing with multiple testing (equipment) platforms, and supporting community-based specimen collection programs. RESULTS Improvements to the NPHL information technology system enabled NPHL to perform >121 000 SARS-CoV-2 polymerase chain reaction tests from March 2020 through January 2022 at a sustainable rate of 2000 SARS-CoV-2 tests per day, with no increase in laboratory staffing. Electronic reporting of 62 000 rapid antigen tests eliminated paper reporting and extended testing services throughout the state. Collection of COVID-19 symptom data before specimen collection enabled NPHL to make data-driven decisions to perform pool testing and conserve testing kits when supplies were low. PRACTICE IMPLICATIONS NPHL information technology applications proved essential for managing health care provider workload, prioritizing the use of scarce testing supplies, and managing Nebraska's overall pandemic response. The NPHL experience provides useful examples of a highly capable information technology system and suggests areas for additional attention in the PHL environment, including a focus on end users, collaboration with various partners, and investment in information technology.
Collapse
Affiliation(s)
- Walter S Campbell
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew Donahue
- Epidemiology Unit, Nebraska Department of Health and Human Services, Lincoln, NE, USA
| | - Robin M Williams
- Epidemiology Unit, Nebraska Department of Health and Human Services, Lincoln, NE, USA
| | - Emily McCutchen
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jana Broadhurst
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andy Schnaubelt
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nicholas J Staffend
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Steven H Hinrichs
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peter C Iwen
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
45
|
Batool R, Soler M, Colavita F, Fabeni L, Matusali G, Lechuga LM. Biomimetic nanoplasmonic sensor for rapid evaluation of neutralizing SARS-CoV-2 monoclonal antibodies as antiviral therapy. Biosens Bioelectron 2023; 226:115137. [PMID: 36796306 PMCID: PMC9904857 DOI: 10.1016/j.bios.2023.115137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/16/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023]
Abstract
Monoclonal antibody (mAb) therapy is one of the most promising immunotherapies that have shown the potential to prevent or neutralize the effects of COVID-19 in patients at very early stages, with a few formulations recently approved by the European and American medicine agencies. However, a main bottleneck for their general implementation resides in the time-consuming, laborious, and highly-specialized techniques employed for the manufacturing and assessing of these therapies, excessively increasing their prices and delaying their administration to the patients. We propose a biomimetic nanoplasmonic biosensor as a novel analytical technique for the screening and evaluation of COVID-19 mAb therapies in a simpler, faster, and reliable manner. By creating an artificial cell membrane on the plasmonic sensor surface, our label-free sensing approach enables real-time monitoring of virus-cell interactions as well as direct analysis of antibody blocking effects in only 15 min assay time. We have achieved detection limits in the 102 TCID50/mL range for the study of SARS-CoV-2 viruses, which allows to perform neutralization assays by only employing a low-volume sample with common viral loads. We have demonstrated the accuracy of the biosensor for the evaluation of two different neutralizing antibodies targeting both Delta and Omicron variants of SARS-CoV-2, with half maximal inhibitory concentrations (IC50) determined in the ng/mL range. Our user-friendly and reliable technology could be employed in biomedical and pharmaceutical laboratories to accelerate, cheapen, and simplify the development of effective immunotherapies for COVID-19 and other serious infectious diseases or cancer.
Collapse
Affiliation(s)
- Razia Batool
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, BIST, CIBER-BBN, Spain
| | - Maria Soler
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, BIST, CIBER-BBN, Spain.
| | - Francesca Colavita
- National Institute for Infectious Disease "L. Spallanzani", IRCCS, Rome, Italy
| | - Lavinia Fabeni
- National Institute for Infectious Disease "L. Spallanzani", IRCCS, Rome, Italy
| | - Giulia Matusali
- National Institute for Infectious Disease "L. Spallanzani", IRCCS, Rome, Italy
| | - Laura M Lechuga
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, BIST, CIBER-BBN, Spain.
| |
Collapse
|
46
|
Kang BH, Jang KW, Yu ES, Na H, Lee YJ, Ko WY, Bae N, Rho D, Jeong KH. Ultrafast Plasmonic Nucleic Acid Amplification and Real-Time Quantification for Decentralized Molecular Diagnostics. ACS NANO 2023; 17:6507-6518. [PMID: 36971140 PMCID: PMC10081571 DOI: 10.1021/acsnano.2c11831] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 03/23/2023] [Indexed: 06/18/2023]
Abstract
Point-of-care real-time reverse-transcription polymerase chain reaction (RT-PCR) facilitates the widespread use of rapid, accurate, and cost-effective near-patient testing that is available to the public. Here, we report ultrafast plasmonic nucleic acid amplification and real-time quantification for decentralized molecular diagnostics. The plasmonic real-time RT-PCR system features an ultrafast plasmonic thermocycler (PTC), a disposable plastic-on-metal (PoM) cartridge, and an ultrathin microlens array fluorescence (MAF) microscope. The PTC provides ultrafast photothermal cycling under white-light-emitting diode illumination and precise temperature monitoring with an integrated resistance temperature detector. The PoM thin film cartridge allows rapid heat transfer as well as complete light blocking from the photothermal excitation source, resulting in real-time and highly efficient PCR quantification. Besides, the MAF microscope exhibits close-up and high-contrast fluorescence microscopic imaging. All of the systems were fully packaged in a palm size for point-of-care testing. The real-time RT-PCR system demonstrates the rapid diagnosis of coronavirus disease-19 RNA virus within 10 min and yields 95.6% of amplification efficiency, 96.6% of classification accuracy for preoperational test, and 91% of total percent agreement for clinical diagnostic test. The ultrafast and compact PCR system can decentralize point-of-care molecular diagnostic testing in primary care and developing countries.
Collapse
Affiliation(s)
- Byoung-Hoon Kang
- Department
of Bio and Brain Engineering, Korea Advanced
Institute of Science and Technology (KAIST), 291 Daehak-ro,
Yuseong-gu, Daejeon 34141, Republic of Korea
- KAIST
Institute for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro,
Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Kyung-Won Jang
- Department
of Bio and Brain Engineering, Korea Advanced
Institute of Science and Technology (KAIST), 291 Daehak-ro,
Yuseong-gu, Daejeon 34141, Republic of Korea
- KAIST
Institute for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro,
Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Eun-Sil Yu
- Department
of Bio and Brain Engineering, Korea Advanced
Institute of Science and Technology (KAIST), 291 Daehak-ro,
Yuseong-gu, Daejeon 34141, Republic of Korea
- KAIST
Institute for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro,
Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Hamin Na
- Department
of Bio and Brain Engineering, Korea Advanced
Institute of Science and Technology (KAIST), 291 Daehak-ro,
Yuseong-gu, Daejeon 34141, Republic of Korea
- KAIST
Institute for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro,
Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Yun-Jae Lee
- OSANG
Healthcare Co., Ltd., 132, Anyangcheondong-ro, Dongan-gu, Anyang-si, Gyeonggi-do 14040, Korea
| | - Woong-Yeol Ko
- OSANG
Healthcare Co., Ltd., 132, Anyangcheondong-ro, Dongan-gu, Anyang-si, Gyeonggi-do 14040, Korea
| | - NamHo Bae
- Center
for Nano Bio Development, National NanoFab
Center (NNFC), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Donggee Rho
- Center
for Nano Bio Development, National NanoFab
Center (NNFC), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Ki-Hun Jeong
- Department
of Bio and Brain Engineering, Korea Advanced
Institute of Science and Technology (KAIST), 291 Daehak-ro,
Yuseong-gu, Daejeon 34141, Republic of Korea
- KAIST
Institute for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro,
Yuseong-gu, Daejeon 34141, Republic of Korea
| |
Collapse
|
47
|
Veroniki AA, Tricco AC, Watt J, Tsokani S, Khan PA, Soobiah C, Negm A, Doherty-Kirby A, Taylor P, Lunny C, McGowan J, Little J, Mallon P, Moher D, Wong S, Dinnes J, Takwoingi Y, Saxinger L, Chan A, Isaranuwatchai W, Lander B, Meyers A, Poliquin G, Straus SE. Rapid antigen-based and rapid molecular tests for the detection of SARS-CoV-2: a rapid review with network meta-analysis of diagnostic test accuracy studies. BMC Med 2023; 21:110. [PMID: 36978074 PMCID: PMC10049780 DOI: 10.1186/s12916-023-02810-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 02/27/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND The global spread of COVID-19 created an explosion in rapid tests with results in < 1 hour, but their relative performance characteristics are not fully understood yet. Our aim was to determine the most sensitive and specific rapid test for the diagnosis of SARS-CoV-2. METHODS Design: Rapid review and diagnostic test accuracy network meta-analysis (DTA-NMA). ELIGIBILITY CRITERIA Randomized controlled trials (RCTs) and observational studies assessing rapid antigen and/or rapid molecular test(s) to detect SARS-CoV-2 in participants of any age, suspected or not with SARS-CoV-2 infection. INFORMATION SOURCES Embase, MEDLINE, and Cochrane Central Register of Controlled Trials, up to September 12, 2021. OUTCOME MEASURES Sensitivity and specificity of rapid antigen and molecular tests suitable for detecting SARS-CoV-2. Data extraction and risk of bias assessment: Screening of literature search results was conducted by one reviewer; data abstraction was completed by one reviewer and independently verified by a second reviewer. Risk of bias was not assessed in the included studies. DATA SYNTHESIS Random-effects meta-analysis and DTA-NMA. RESULTS We included 93 studies (reported in 88 articles) relating to 36 rapid antigen tests in 104,961 participants and 23 rapid molecular tests in 10,449 participants. Overall, rapid antigen tests had a sensitivity of 0.75 (95% confidence interval 0.70-0.79) and specificity of 0.99 (0.98-0.99). Rapid antigen test sensitivity was higher when nasal or combined samples (e.g., combinations of nose, throat, mouth, or saliva samples) were used, but lower when nasopharyngeal samples were used, and in those classified as asymptomatic at the time of testing. Rapid molecular tests may result in fewer false negatives than rapid antigen tests (sensitivity: 0.93, 0.88-0.96; specificity: 0.98, 0.97-0.99). The tests with the highest sensitivity and specificity estimates were the Xpert Xpress rapid molecular test by Cepheid (sensitivity: 0.99, 0.83-1.00; specificity: 0.97, 0.69-1.00) among the 23 commercial rapid molecular tests and the COVID-VIRO test by AAZ-LMB (sensitivity: 0.93, 0.48-0.99; specificity: 0.98, 0.44-1.00) among the 36 rapid antigen tests we examined. CONCLUSIONS Rapid molecular tests were associated with both high sensitivity and specificity, while rapid antigen tests were mainly associated with high specificity, according to the minimum performance requirements by WHO and Health Canada. Our rapid review was limited to English, peer-reviewed published results of commercial tests, and study risk of bias was not assessed. A full systematic review is required. REVIEW REGISTRATION PROSPERO CRD42021289712.
Collapse
Affiliation(s)
- Areti Angeliki Veroniki
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, 209 Victoria Street, East Building, Toronto, ON M5B 1T8 Canada
- Institute for Health Policy, Management, and Evaluation, University of Toronto, Toronto, ON Canada
| | - Andrea C. Tricco
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, 209 Victoria Street, East Building, Toronto, ON M5B 1T8 Canada
- Epidemiology Division & Institute of Health Policy, Management, and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON Canada
- Queen’s Collaboration for Health Care Quality: A JBI Centre of Excellence, Kingston, Canada
| | - Jennifer Watt
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, 209 Victoria Street, East Building, Toronto, ON M5B 1T8 Canada
| | - Sofia Tsokani
- School of Education, University of Ioannina, Ioannina, Greece
| | - Paul A. Khan
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, 209 Victoria Street, East Building, Toronto, ON M5B 1T8 Canada
| | - Charlene Soobiah
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, 209 Victoria Street, East Building, Toronto, ON M5B 1T8 Canada
- Institute for Health Policy, Management, and Evaluation, University of Toronto, Toronto, ON Canada
| | - Ahmed Negm
- University of Alberta, Edmonton, AB Canada
| | - Amanda Doherty-Kirby
- Patient Partner, Strategy for Patient Oriented-Research Evidence Alliance (SPOR EA), Toronto, Canada
| | - Paul Taylor
- Patient Partner, Strategy for Patient Oriented-Research Evidence Alliance (SPOR EA), Toronto, Canada
| | - Carole Lunny
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, 209 Victoria Street, East Building, Toronto, ON M5B 1T8 Canada
| | - Jessie McGowan
- University of Ottawa/Université d’Ottawa, Ottawa, ON Canada
| | - Julian Little
- University of Ottawa/Université d’Ottawa, Ottawa, ON Canada
| | | | - David Moher
- Ottawa Hospital Research Institute/Institut de Recherche de L’Hôpital d’Ottawa, Ottawa, ON Canada
| | - Sabrina Wong
- University of British Columbia, Vancouver, BC Canada
| | | | | | | | | | | | - Bryn Lander
- Health Canada (Ottawa)/Santé Canada (Ottawa), Ottawa, ON Canada
| | - Adrienne Meyers
- Public Health Agency of Canada/Agence de La Santé Publique du Canada, Ottawa, ON Canada
| | - Guillaume Poliquin
- Public Health Agency of Canada/Agence de La Santé Publique du Canada, Ottawa, ON Canada
| | - Sharon E. Straus
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, 209 Victoria Street, East Building, Toronto, ON M5B 1T8 Canada
- Institute for Health Policy, Management, and Evaluation, University of Toronto, Toronto, ON Canada
- Department of Geriatric Medicine, University of Toronto, Toronto, ON Canada
| |
Collapse
|
48
|
Jang HJ, Zhuang W, Sui X, Ryu B, Huang X, Chen M, Cai X, Pu H, Beavis K, Huang J, Chen J. Rapid, Sensitive, Label-Free Electrical Detection of SARS-CoV-2 in Nasal Swab Samples. ACS APPLIED MATERIALS & INTERFACES 2023; 15:15195-15202. [PMID: 36938607 PMCID: PMC10041344 DOI: 10.1021/acsami.3c00331] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/07/2023] [Indexed: 06/18/2023]
Abstract
Rapid diagnosis of coronavirus disease 2019 (COVID-19) is key for the long-term control of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) amid renewed threats of mutated SARS-CoV-2 around the world. Here, we report on an electrical label-free detection of SARS-CoV-2 in nasopharyngeal swab samples directly collected from outpatients or in saliva-relevant conditions by using a remote floating-gate field-effect transistor (RFGFET) with a 2-dimensional reduced graphene oxide (rGO) sensing membrane. RFGFET sensors demonstrate rapid detection (<5 min), a 90.6% accuracy from 8 nasal swab samples measured by 4 different devices for each sample, and a coefficient of variation (CV) < 6%. Also, RFGFET sensors display a limit of detection (LOD) of pseudo-SARS-CoV-2 that is 10 000-fold lower than enzyme-linked immunosorbent assays, with a comparable LOD to that of reverse transcription-polymerase chain reaction (RT-PCR) for patient samples. To achieve this, comprehensive systematic studies were performed regarding interactions between SARS-CoV-2 and spike proteins, neutralizing antibodies, and angiotensin-converting enzyme 2, as either a biomarker (detection target) or a sensing probe (receptor) functionalized on the rGO sensing membrane. Taken together, this work may have an immense effect on positioning FET bioelectronics for rapid SARS-CoV-2 diagnostics.
Collapse
Affiliation(s)
- Hyun-June Jang
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
- Chemical
Sciences and Engineering Division, Physical Sciences and Engineering
Directorate, Argonne National Laboratory, Lemont, Illinois 60439, United States
| | - Wen Zhuang
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
- Chemical
Sciences and Engineering Division, Physical Sciences and Engineering
Directorate, Argonne National Laboratory, Lemont, Illinois 60439, United States
| | - Xiaoyu Sui
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
- Chemical
Sciences and Engineering Division, Physical Sciences and Engineering
Directorate, Argonne National Laboratory, Lemont, Illinois 60439, United States
| | - Byunghoon Ryu
- Chemical
Sciences and Engineering Division, Physical Sciences and Engineering
Directorate, Argonne National Laboratory, Lemont, Illinois 60439, United States
| | - Xiaodan Huang
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Min Chen
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Xiaolei Cai
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Haihui Pu
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
- Chemical
Sciences and Engineering Division, Physical Sciences and Engineering
Directorate, Argonne National Laboratory, Lemont, Illinois 60439, United States
| | - Kathleen Beavis
- Department
of Pathology, University of Chicago, Chicago, Illinois 60637, United States
| | - Jun Huang
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Junhong Chen
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
- Chemical
Sciences and Engineering Division, Physical Sciences and Engineering
Directorate, Argonne National Laboratory, Lemont, Illinois 60439, United States
| |
Collapse
|
49
|
Krishnamoorthy A, Chandrapalan S, JalayeriNia G, Hussain Y, Bannaga A, Lei II, Arasaradnam R. Influence of seasonal and operator variations on diagnostic accuracy of lateral flow devices during the COVID-19 pandemic: a systematic review and meta-analysis. Clin Med (Lond) 2023; 23:144-150. [PMID: 36921988 PMCID: PMC11046551 DOI: 10.7861/clinmed.2022-0319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
BACKGROUND Lateral flow tests (LFT) are point-of-care rapid antigen tests that allow isolation and control of disease outbreaks through convenient, practical testing. However, studies have shown significant variation in their diagnostic accuracy. We conducted a systematic review of the diagnostic accuracy of LFTs for the detection of severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) to identify potential factors affecting their performance. METHODS A systematic search of online databases was carried out to identify studies assessing the sensitivity and specificity of LFTs compared with polymerase chain reaction (PCR) tests. Data were extracted and used to calculate pooled sensitivity and specificity. Meta-regression analysis was conducted to identify covariates influencing diagnostic accuracy. RESULTS In total, 76 articles with 108,820 test results were identified for analysis. Pooled sensitivity and specificity were 72% (95% confidence interval (CI): 0.68-0.76) and 100% (95% CI: 0.99-1.00), respectively. Staff operation of the LFT showed a statistically significant increase in sensitivity (p=0.04) and specificity (p=0.001) compared with self-operation by the test subjects. The use of LFTs in symptomatic patient subgroups also resulted in higher test sensitivity. CONCLUSION LFTs display good sensitivity and extremely good specificity for SARS-CoV-2 antigen detection; they become more sensitive in patients with symptoms and when performed by trained professionals.
Collapse
Affiliation(s)
| | | | - Gohar JalayeriNia
- University Hospital of Coventry & Warwickshire NHS Trust, Coventry, UK
| | - Yaqza Hussain
- University Hospital of Coventry & Warwickshire NHS Trust, Coventry, UK
| | - Ayman Bannaga
- University Hospital of Coventry & Warwickshire NHS Trust, Coventry, UK
| | - Ian Io Lei
- University Hospital of Coventry & Warwickshire NHS Trust, Coventry, UK
| | | |
Collapse
|
50
|
Ortiz-Millán G. COVID-19 Health Passes: Practical and Ethical Issues. JOURNAL OF BIOETHICAL INQUIRY 2023; 20:125-138. [PMID: 36630062 PMCID: PMC9832398 DOI: 10.1007/s11673-022-10227-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 08/28/2022] [Indexed: 05/04/2023]
Abstract
Several countries have implemented COVID-19 health passes or certificates to promote a safer return to in-person social activities. These passes have been proposed as a way to prove that someone has been vaccinated, has recovered from the disease, or has negative results on a diagnostic test. However, many people have questioned their ethical justification. This article presents some practical and ethical problems to consider in the event of wishing to implement these passes. Among the former, it is questioned how accurate diagnostic tests are as a means of ensuring that a person is not contagious, whether vaccination guarantees immunity, the fact that health passes can be forged, whether they encourage vaccination, and the problem that there is no universally recognized health pass. Among the ethical issues, it is discussed whether health passes promote discrimination and inequality and whether they violate rights to privacy and freedom. It is concluded that health passes have enough ethical justification to be implemented.
Collapse
Affiliation(s)
- Gustavo Ortiz-Millán
- Instituto de Investigaciones Filosóficas, Universidad Nacional Autónoma de México (UNAM), Circuito Mario de la Cueva s/n, Ciudad Universitaria, Coyoacán, 04510, Mexico City, Mexico.
| |
Collapse
|