1
|
Sathyanarayana SH, Robins AA, Toledo DM, Gallagher TL, Tsongalis GJ, Hubbard JA, Lefferts JA, Martin IW. Simplifying SARS-CoV-2 wastewater-based surveillance using an automated FDA EUA assay. Microbiol Spectr 2025; 13:e0249024. [PMID: 39998239 PMCID: PMC11960137 DOI: 10.1128/spectrum.02490-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/08/2025] [Indexed: 02/26/2025] Open
Abstract
Wastewater-based surveillance (WBS) can track the spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in communities. Laboratory methods for this testing involve labor-intensive, multi-step processes. This study assessed the feasibility of performing WBS with an off-label use of an automated commercial SARS-CoV-2 assay that had received Emergency Use Authorization for human diagnostic testing from the United States Food and Drug Administration (FDA EUA). Twenty-four-hour composite samples of primary influent wastewater from seven municipalities in New Hampshire and Vermont were collected between September 2020 and February 2021, and were centrifuged upon receipt. An aliquot of fresh supernatant was immediately tested with the Abbott m2000 RealTime SARS-CoV-2 assay (Abbott Molecular, Des Plaines, IL, USA). Corresponding aliquots were then stored at -80°C until they were thawed, polyethylene glycol (PEG) concentrated, and tested by two PCR-based laboratory-developed tests (LDTs). Wastewater samples (103) were tested with successful detection of SARS-CoV-2 viral RNA by all three methods. Bland-Altman analysis showed overall concordant results with a bias of -0.13 and -0.42 log copies/mL detected by the FDA EUA assay compared to the LDTs. Specimen stability assessment demonstrated a decrease of 33.9% measurable viral RNA after three freeze-thaw cycles. SARS-CoV-2 detection in wastewater using an FDA EUA assay on an automated commercial testing platform performed comparably but with more efficient workflow when compared to two LDTs. This sample-to-answer automated method could save time and labor for surveillance testing, but further validation of its ability to quantitate SARS-CoV-2 viral RNA is necessary.IMPORTANCEThis proof-of-principle study evaluates an off-label use of an automated United States Food and Drug Administration (FDA) Emergency Use Authorization (EUA) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) human diagnostic assay for wastewater surveillance. Compared to standard, labor-intensive, multi-step methods currently in use for wastewater surveillance testing, an off-label use of an FDA EUA assay on an automated platform offers a sample-to-answer testing requiring less labor and a faster turnaround time.
Collapse
Affiliation(s)
| | - Ashlee A. Robins
- Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire, USA
| | - Diana M. Toledo
- The Broad Institute at MIT and Harvard, Cambridge, Massachusetts, USA
| | - Torrey L. Gallagher
- Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire, USA
| | - Gregory J. Tsongalis
- Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire, USA
| | | | - Joel A. Lefferts
- Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire, USA
| | - Isabella W. Martin
- Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire, USA
| |
Collapse
|
2
|
Crawford KHD, Baniecki ML, Dushin EG, Tierney CA, Guan S, Stensland LL, Perez-Osorio AC, Greninger AL. Specimen adequacy assay controls in nucleic acid amplification tests do not correlate with nasopharyngeal swab collection method. J Clin Microbiol 2024; 62:e0097524. [PMID: 39283073 PMCID: PMC11481563 DOI: 10.1128/jcm.00975-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2024] Open
Affiliation(s)
- Katharine H. D. Crawford
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | | | | | | | | | - Laurence L. Stensland
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Ailyn C. Perez-Osorio
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Alexander L. Greninger
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Research Center, Seattle, Washington, USA
| |
Collapse
|
3
|
Stojčević-Maletić J, Barjaktarović I, Đerić M, Čabarkapa V, Petrović V, Patić A, Bogdanović-Vasić S. COMPARISON OF ABBOTT REAL TIME SARS-COV-2 ASSAY AND LIFERIVER NOVEL CORONAVIRUS REAL TIME MULTIPLEX KIT FOR THE RT-PCR BASED DETECTION OF SARS-COV-2 FROM NASOPHARYNGEAL SWABS. Acta Clin Croat 2024; 63:275-282. [PMID: 40104229 PMCID: PMC11912847 DOI: 10.20471/acc.2024.63.02.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 02/15/2022] [Indexed: 03/20/2025] Open
Abstract
The objective of this study was to compare performance of two authorized tests, the Abbott Real Time SARS-CoV-2 (ACOV) assay (Abbott Molecular Inc., North Chicago, IL, USA) and Liferiver Novel Coronavirus Real Time Multiplex RT-PCR (Liferiver 2019-nCoV) kit (BioVendor Group, Brno, Czech Republic), and to determine whether the selection of targeted genes has an impact on test specificity. We included 105 nasopharyngeal swabs from adult individuals with symptoms or suspected of coronavirus disease 2019 (COVID-19), aged from 26 to 91 years, previously tested by the ACOV and subsequently tested by the Liferiver 2019-nCoV. In this comparative analysis, we found that the ACOV assay detected more cases of COVID-19 infection than the Liferiver 2019-nCoV assay. The Liferiver 2019-nCoV kit showed a high level of agreement with the ACOV assay. The positive percent agreement was 88.89% (95% confidence intervals (95% CI): 77.42%-100.0%), and the kappa coefficient (kappa) was 0.901 (95% CI: 0.817-0.985). The negative percent agreement was 94.12% (95% CI: 89.74%-98.50%), while 4.76% of SARS-CoV-2 cases were false-negative using the Liferiver test. However, due to the possible false-negative results using the Liferiver 2019-nCOV test, we recommend complete testing with the ACOV assay.
Collapse
Affiliation(s)
- Jelena Stojčević-Maletić
- University of Novi Sad, Faculty of Medicine, Novi Sad, Serbia
- University Clinical Center of Vojvodina, Center for Laboratory Diagnostics, Novi Sad, Serbia
| | - Iva Barjaktarović
- University of Novi Sad, Faculty of Medicine, Novi Sad, Serbia
- University Clinical Center of Vojvodina, Center for Laboratory Diagnostics, Novi Sad, Serbia
| | - Mirjana Đerić
- University of Novi Sad, Faculty of Medicine, Novi Sad, Serbia
- University Clinical Center of Vojvodina, Center for Laboratory Diagnostics, Novi Sad, Serbia
| | - Velibor Čabarkapa
- University of Novi Sad, Faculty of Medicine, Novi Sad, Serbia
- University Clinical Center of Vojvodina, Center for Laboratory Diagnostics, Novi Sad, Serbia
| | - Vladimir Petrović
- University of Novi Sad, Faculty of Medicine, Novi Sad, Serbia
- Institute of Public Health of Vojvodina, Novi Sad, Serbia
| | - Aleksandra Patić
- University of Novi Sad, Faculty of Medicine, Novi Sad, Serbia
- Institute of Public Health of Vojvodina, Novi Sad, Serbia
| | | |
Collapse
|
4
|
Paul MK, Choudhary MC, Heaps AL, Deo R, Moisi D, Gordon KC, Mellors JW, Moser C, Klekotka P, Landay A, Currier JS, Eron JJ, Chew KW, Smith DM, Sieg SF, Parikh UM, Li JZ. Impact of SARS-CoV-2 Resistance to Antiviral Monoclonal Antibody Therapy on Neutralizing Antibody Response. Pathog Immun 2024; 9:79-93. [PMID: 39247686 PMCID: PMC11378757 DOI: 10.20411/pai.v9i2.718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/17/2024] [Indexed: 09/10/2024] Open
Abstract
Background Anti-SARS-CoV-2 monoclonal antibodies (mAbs) have played a key role as an anti-viral against SARS-CoV-2, but there is a potential for resistance to develop. The interplay between host antibody responses and the development of monoclonal antibody (mAb) resistance is a critical area of investigation. In this study, we assessed host neutralizing antibody (nAb) responses against both ancestral virus and those with treatment-emergent E484K bamlanivimab resistance mutations. Methods Study participants were enrolled in the ACTIV-2/Advancing Clinical Therapeutics Globally (ACTG) A5401 phase 2 randomized, placebo-controlled trial of bamlanivimab 700 mg mAb therapy (NCT04518410). Anterior nasal and nasopharyngeal swabs were collected for SARS-CoV-2 RNA testing and S gene next-generation sequencing to identify the E484K bamlanivimab resistance mutation. Serum nAb titers were assessed by pseudovirus neutralization assays. Results Higher baseline (pre-treatment) nAb titers against either ancestral or E484K virus was associated with lower baseline viral load. Participants with emerging resistance had low levels of nAb titers against either ancestral or E484K nAb at the time of study entry. Participants with emergent E484K resistance developed significantly higher levels of E484K-specific nAb titers compared to mAb-treated individuals who did not develop resistance. All participants who developed the E484K mAb resistance mutation were eventually able to clear the virus. Conclusion Emerging drug resistance after SARS-CoV-2-specific mAb therapy led to a heightened host neutralizing antibody response to the mAb-resistant variant that was associated with eventual viral clearance. This demonstrates the interplay between the antiviral treatment-directed viral evolution and subsequent host immune response in viral clearance.
Collapse
Affiliation(s)
- Marc-Kendy Paul
- Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA
| | | | - Amy L. Heaps
- University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Rinki Deo
- Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA
| | | | | | | | - Carlee Moser
- Harvard T.H. Chan School of Public Health, Boston, MA
| | | | | | - Judith S. Currier
- David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Joseph J. Eron
- University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Kara W. Chew
- David Geffen School of Medicine, University of California, Los Angeles, CA
| | | | - Scott F. Sieg
- Case Western Reserve University and University Hospitals Cleveland, Cleveland, OH
| | - Urvi M. Parikh
- University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jonathan Z. Li
- Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA
| | - on behalf of the ACTIV-2/A5401 Study Team
- Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA
- University of Pittsburgh School of Medicine, Pittsburgh, PA
- Case Western Reserve University, Cleveland, OH
- Harvard T.H. Chan School of Public Health, Boston, MA
- Eli Lilly and Company, San Diego, CA
- RUSH Medical College, Chicago, IL
- David Geffen School of Medicine, University of California, Los Angeles, CA
- University of North Carolina at Chapel Hill, Chapel Hill, NC
- University of California, San Diego, CA
- Case Western Reserve University and University Hospitals Cleveland, Cleveland, OH
| |
Collapse
|
5
|
Choudhary MC, Deo R, Evering TH, Chew KW, Giganti MJ, Moser C, Ritz J, Regan J, Flynn JP, Crain CR, Wohl DA, Currier JS, Eron JJ, Margolis D, Zhu Q, Zhon L, Ya L, Greninger AL, Hughes MD, Smith D, Daar ES, Li JZ. Characterization of Treatment Resistance and Viral Kinetics in the Setting of Single-Active Versus Dual-Active Monoclonal Antibodies Against Severe Acute Respiratory Syndrome Coronavirus 2. J Infect Dis 2024; 230:394-402. [PMID: 38716969 PMCID: PMC11326811 DOI: 10.1093/infdis/jiae192] [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: 03/15/2024] [Accepted: 04/11/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Monoclonal antibodies (mAbs) represent a crucial antiviral strategy for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, but it is unclear whether combination mAbs offer a benefit over single-active mAb treatment. Amubarvimab and romlusevimab significantly reduced the risk of hospitalizations or death in the ACTIV-2/A5401 trial. Certain SARS-CoV-2 variants are intrinsically resistant against romlusevimab, leading to only single-active mAb therapy with amubarvimab in these variants. We evaluated virologic outcomes in individuals treated with single- versus dual-active mAbs. METHODS Participants were nonhospitalized adults at higher risk of clinical progression randomized to amubarvimab plus romlusevimab or placebo. Quantitative SARS-CoV-2 RNA levels and targeted S-gene next-generation sequencing was performed on anterior nasal samples. We compared viral load kinetics and resistance emergence between individuals treated with effective single- versus dual-active mAbs depending on the infecting variant. RESULTS Study participants receiving single- or dual-active mAbs had similar demographics, baseline nasal viral load, symptom score, and symptom duration. Compared with single-active mAb treatment, treatment with dual-active mAbs led to faster viral load decline at study days 3 (P < .001) and 7 (P < .01). Treatment-emergent resistance mutations were more likely to be detected after amubarvimab plus romlusevimab treatment than with placebo (2.6% vs 0%; P < .001) and were more frequently detected in the setting of single-active compared with dual-active mAb treatment (7.3% vs 1.1%; P < .01). Single-active and dual-active mAb treatment resulted in similar decrease in rates of hospitalizations or death. CONCLUSIONS Compared with single-active mAb therapy, dual-active mAbs led to similar clinical outcomes but significantly faster viral load decline and a lower risk of emergent resistance.
Collapse
Affiliation(s)
- Manish C Choudhary
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rinki Deo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Teresa H Evering
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Kara W Chew
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Mark J Giganti
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Carlee Moser
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Justin Ritz
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - James Regan
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James P Flynn
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Charles R Crain
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David Alain Wohl
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Judith S Currier
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Joseph J Eron
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | | | - Qing Zhu
- Brii Biosciences, Durham, North Carolina, USA
| | - Lijie Zhon
- Brii Biosciences, Durham, North Carolina, USA
| | - Li Ya
- Brii Biosciences, Durham, North Carolina, USA
| | - Alexander L Greninger
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Michael D Hughes
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Davey Smith
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Eric S Daar
- Department of Medicine, Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Jonathan Z Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Theodorsson E, Meijer P, Badrick T. External quality assurance in the era of standardization. Clin Chim Acta 2024; 557:117876. [PMID: 38493945 DOI: 10.1016/j.cca.2024.117876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Metrology in clinical chemistry aims to ensure the equivalence of measurement results from different in-vitro diagnostic measurement devices (IVD MD) for use in healthcare. The metrological traceability of measurement results to higher-order references is the cornerstone to achieving equivalent results. However, other fundamentals are also needed, including the commutability of reference materials and external quality assessment (EQA) materials for monitoring the equivalence of measurement results at the end-user level. This manuscript summarizes the findings and opinions expressed at the Joint Community for Traceability in Laboratory Medicine (JCTLM) workshop held on December 4-5, 2023. The workshop explored the relationship between EQA/proficiency testing and metrological traceability to higher-order references. EQA monitors the equivalence of measurement results from end-user IVD MDs. The workshop discussed the role and challenges of using EQA to improve and maintain the equivalence of measurement results. It also elucidated current developments in establishing the clinical suitability of laboratory results expressed as analytical performance specifications (APS).
Collapse
Affiliation(s)
- Elvar Theodorsson
- Department of Biomedical and Clinical Sciences, Division of Clinical Chemistry and Pharmacology, Linköping University, Linköping, Sweden.
| | - Piet Meijer
- ECAT Foundation, Voorschoten, The Netherlands
| | - Tony Badrick
- RCPA Quality Assurance Programs, St Leonards, Sydney, Australia
| |
Collapse
|
7
|
Chung PYJ, Dhillon SK, Simoens C, Cuypers L, Laenen L, Bonde J, Corbisier P, Buttinger G, Cocuzza CE, Van Gucht S, Van Ranst M, Arbyn M. Assessment of the clinical and analytical performance of three Seegene Allplex SARS-CoV-2 assays within the VALCOR framework. Microbiol Spectr 2024; 12:e0239723. [PMID: 38189291 PMCID: PMC10846132 DOI: 10.1128/spectrum.02397-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic demonstrated the need for accurate diagnostic testing for the early detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although the pandemic has ended, accurate assays are still needed to monitor viral spread at national levels and beyond through population and wastewater surveillance. To enhance early detection, SARS-CoV-2 assays should have high diagnostic accuracy and should be validated to assure accurate results. Three distinct SARS-CoV-2 assays were evaluated with clinical samples using the VALCOR (VALidation of SARS-CORona Virus-2 assays) framework, with the TaqPath COVID-19 assay (ThermoFisher Scientific, USA) as a comparator. We evaluated clinical sensitivity, specificity, limit of detection (LOD), and overall concordance between comparator and three index Allplex SARS-CoV-2 assays (Seegene, South Korea): Allplex-SC2, Allplex-SC2Fast (Fast PCR), and Allplex-SC2FabR (SARS-CoV-2/FluA/FluB/respiratory syncytial virus). Analytical performance and LOD of index assays were assessed using a dilution series of three synthetic SARS-CoV-2 sequence reference materials (RMs). Ninety SARS-CoV-2 positives and 90 SARS-CoV-2 negatives were tested. All Allplex assays had 100.0% sensitivity (95%CI = 95.9%-100.0%). Allplex-SC2 and Allplex-SC2Fast assays had 97.8% specificity (95%CI = 92.3%-99.7%) and 98.9% overall concordance [κ = 0.978 (95%CI = 0.947-1.000)]. Allplex-SC2FabR assay showed 100.0% specificity (95%CI = 95.9%-100.0%) and 100.0% overall concordance [κ = 1.000 (95%CI = 1.000-1.000)]. LOD assessment of index assays revealed detection down to 2.61 × 102 copies/mL in clinical samples, while the analytical LOD was 9.00 × 102 copies/mL. In conclusion, the evaluation of the three Seegene Allplex SARS-CoV-2 assays showed high sensitivity and specificity and an overall good assay concordance with the comparator. The assays showed low analytical LOD using RM and even a slightly lower LOD in clinical samples. Non-overlapping target gene sequences between SARS-CoV-2 assays and RMs emphasize the need for aligning targeted sequences of diagnostic assays and RMs.IMPORTANCEThe coronavirus disease 2019 pandemic has a significant impact on global public health, economies, and societies. As shown through the first phases of the pandemic, accurate and timely diagnosis is crucial for disease control, prevention, and monitoring. Though the pandemic phase of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has concluded, diagnostic assays remain in demand to monitor SARS-CoV-2 at the individual patient level, regionally, and nationally, as well as to remain an infectious disease preparedness instrument to monitor any new SARS-CoV-2 dissemination across borders using population and wastewater surveillance. The anticipation by WHO and central health care policy entities such as the Center for Disease Control, EMA, and multiple national health authorities is that SARS-CoV-2 will reside as an endemic respiratory disease for years to come. The key strategic consideration is hence shifting from combating a pandemic situation with a high number of patients to instead allowing precise diagnostics of suspected patients with the intention of correct management in a low-prevalence setting.
Collapse
Affiliation(s)
- Pui Yan Jenny Chung
- Unit of Cancer Epidemiology, Belgian Cancer Centre, Sciensano, Brussels, Belgium
| | - Sharonjit K. Dhillon
- Unit of Cancer Epidemiology, Belgian Cancer Centre, Sciensano, Brussels, Belgium
| | - Cindy Simoens
- Unit of Cancer Epidemiology, Belgian Cancer Centre, Sciensano, Brussels, Belgium
| | - Lize Cuypers
- Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Clinical Microbiology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Lies Laenen
- Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Clinical Microbiology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jesper Bonde
- Molecular Pathology Laboratory, Department of Pathology, AHH-Hvidovre Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Clementina E. Cocuzza
- Laboratory of Clinical Microbiology and Virology, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Marc Van Ranst
- Laboratory of Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Marc Arbyn
- Unit of Cancer Epidemiology, Belgian Cancer Centre, Sciensano, Brussels, Belgium
- Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, University of Ghent, Ghent, Belgium
| |
Collapse
|
8
|
Hafez W, Nasa P, Rashid A, Saleh H, Abuelsaoud HM, Abdelshakour M, Nashef Z, Abazid RR, Ahmed AB, Ahmed S, Gador M, Faheem Y, John S, Mohamed AA, Abdoh A. Interleukin 6 (IL6) as a potential interplaying factor between obesity and COVID-19 outcome. INFORMATICS IN MEDICINE UNLOCKED 2024; 45:101455. [DOI: 10.1016/j.imu.2024.101455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
|
9
|
Babic N, Garner KS, Hirschhorn JW, Zebian R, Nolte FS. Evaluation of Abbott ID NOW COVID-19 POC test performance characteristics and integration in the regional health network workflows to improve health care delivery. Clin Biochem 2023; 117:69-73. [PMID: 34896098 PMCID: PMC8653396 DOI: 10.1016/j.clinbiochem.2021.12.003] [Citation(s) in RCA: 2] [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: 08/31/2021] [Revised: 10/18/2021] [Accepted: 12/03/2021] [Indexed: 12/01/2022]
Abstract
With the recent global surge of SARS-CoV-2 Delta variant, there continues to be high demand for COVID-19 diagnostic testing. Abbott ID NOW is a rapid, CLIA-waived, COVID-19 diagnostic test ideally suited for use in urgent care settings or where access to diagnostic testing is limited. In this study we describe the results of rigorous validation of ID NOW and post-implementation study of POC test utilization patterns within community hospitals and clinics. Performance of ID NOW was validated by comparison of the results from 207 consecutive, paired, specimens tested on the ID NOW and on the m2000/Alinity m platforms. Once validated, ID NOW devices were placed for clinical use at four regional hospitals and clinics. We found that the ID NOW and m2000/Alinity m positive and negative percent agreement were 94.5% (95% CI, 85.1% to 98.1%) and 99.3% (95% CI, 96.4% to 99.9%), respectively. As of August 2021, a total of 2,301 tests were performed by ID NOW at individual regional network sites. The population tested consisted of 55.5% White and 42.9% Black patients, with Black patients presenting predominantly in the hospitals, while White patients were more evenly distributed between hospital and clinic sites. Disease prevalence observed among patients tested by ID NOW (12.3%) was aligned with overall prevalence seen at regional sites (11.3%). In summary, the ID NOW test can provide rapid and accurate results in a variety of near-to-patient and POC settings. If used correctly, it could serve as a valuable diagnostic tool to enable equal access to care and improve healthcare delivery within large health network systems.
Collapse
Affiliation(s)
- N Babic
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, United States.
| | - K S Garner
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, United States
| | - J W Hirschhorn
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, United States
| | - R Zebian
- Department of Pulmonary and Critical Care Medicine, MUSC Health Florence Division, Florence, SC 29505, United States
| | - F S Nolte
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, United States
| |
Collapse
|
10
|
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: 23] [Impact Index Per Article: 11.5] [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
|
11
|
Romano ER, Sleeman K, Hall-Eidson P, Zeh C, Bhairavabhotla R, Zhang G, Adhikari A, Alemnji G, Cardo YR, Pinheiro A, Pocongo B, Eno LT, Shang JD, Ndongmo CB, Rosario H, Moreno O, De León LADLC, Fonjungo P, Kabwe C, Ahuke-Mundeke S, Gama D, Dlamini S, Maphalala G, Abreha T, Purfield A, Gebrehiwot YT, Desalegn DM, Basiye F, Mwangi J, Bowen N, Mengistu Y, Lecher S, Kampira E, Kaba M, Bitilinyu-Bangoh J, Masamha G, Viegas SO, Beard RS, van Rooyen G, Shiningavamwe AN, I J M, Iriemenam NC, Mba N, Okoi C, Katoro J, Kenyi DL, Bior BK, Mwangi C, Nabadda S, Kaleebu P, Yingst SL, Chikwanda P, Veri L, Simbi R, Alexander H. Contribution of PEPFAR-Supported HIV and TB Molecular Diagnostic Networks to COVID-19 Testing Preparedness in 16 Countries. Emerg Infect Dis 2022; 28:S59-S68. [PMID: 36502414 PMCID: PMC9745242 DOI: 10.3201/eid2813.220789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The US President's Emergency Plan for AIDS Relief (PEPFAR) supports molecular HIV and tuberculosis diagnostic networks and information management systems in low- and middle-income countries. We describe how national programs leveraged these PEPFAR-supported laboratory resources for SARS-CoV-2 testing during the COVID-19 pandemic. We sent a spreadsheet template consisting of 46 indicators for assessing the use of PEPFAR-supported diagnostic networks for COVID-19 pandemic response activities during April 1, 2020, to March 31, 2021, to 27 PEPFAR-supported countries or regions. A total of 109 PEPFAR-supported centralized HIV viral load and early infant diagnosis laboratories and 138 decentralized HIV and TB sites reported performing SARS-CoV-2 testing in 16 countries. Together, these sites contributed to >3.4 million SARS-CoV-2 tests during the 1-year period. Our findings illustrate that PEPFAR-supported diagnostic networks provided a wide range of resources to respond to emergency COVID-19 diagnostic testing in 16 low- and middle-income countries.
Collapse
|
12
|
Hailemariam BW, Zealiyas K, Gutema G, Gebremicael G, Adane S, Tadele S, Tayachew A, Araya S, Desta K. Performances of four Nucleic Acid Amplification Tests for the identification of SARS-CoV-2 in Ethiopia. Sci Rep 2022; 12:20282. [PMID: 36434013 PMCID: PMC9700788 DOI: 10.1038/s41598-022-24411-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022] Open
Abstract
Since Coronavirus Disease-2019 (COVID-19) outbreak was reported, many commercial Nucleic Acid Amplification Tests (NAAT) have been developed all over the world, and it has been the standard method. Even though several assays were rapidly developed and applied to laboratory diagnostic testing, the performance of these assays was not evaluated in different contexts. Thus, this study aimed to assess the performance of Abbott SARS-CoV-2, Daan Gene, BGI and Sansure Biotech assays by using Composite Reference Standard (CRS). The study was conducted at the Ethiopian Public Health Institute (EPHI) from December 1 to 30/2020. Of the 164 nasopharyngeal samples were extracted by using a QIAamp RNA mini kit and Abbott DNA sample preparation system. Out of 164 samples, 59.1% were positive and 40.9% were negative by CRS. Sansure Biotech positivity was significantly low compared to CRS (p < 0.05). The overall agreement of the four assays compared to CRS was 96.3-100%. The performance of the four assays had almost comparable diagnostic performance, except for a low positive rate of Sansure Biotech assay. Hence, Sansure Biotech assay [Research Use Only (RUO)] needs further verification on its use in Ethiopia. Finally an additional study should be considered for evaluating assays with respective manufacturer claims.
Collapse
Affiliation(s)
- Belete Woldesemayat Hailemariam
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia.
- Department of Medical Laboratory Sciences, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia.
| | - Kidist Zealiyas
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Gadissa Gutema
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Gebremedhin Gebremicael
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Sisay Adane
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Sisay Tadele
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Adamu Tayachew
- National Influenza Reference Laboratory, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Shambel Araya
- Department of Medical Laboratory Sciences, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Kassu Desta
- Department of Medical Laboratory Sciences, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
13
|
Choudhary MC, Chew KW, Deo R, Flynn JP, Regan J, Crain CR, Moser C, Hughes MD, Ritz J, Ribeiro RM, Ke R, Dragavon JA, Javan AC, Nirula A, Klekotka P, Greninger AL, Fletcher CV, Daar ES, Wohl DA, Eron JJ, Currier JS, Parikh UM, Sieg SF, Perelson AS, Coombs RW, Smith DM, Li JZ. Emergence of SARS-CoV-2 escape mutations during Bamlanivimab therapy in a phase II randomized clinical trial. Nat Microbiol 2022; 7:1906-1917. [PMID: 36289399 PMCID: PMC9675946 DOI: 10.1038/s41564-022-01254-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 09/19/2022] [Indexed: 11/08/2022]
Abstract
SARS-CoV-2 mutations that cause resistance to monoclonal antibody (mAb) therapy have been reported. However, it remains unclear whether in vivo emergence of SARS-CoV-2 resistance mutations alters viral replication dynamics or therapeutic efficacy in the immune-competent population. As part of the ACTIV-2/A5401 randomized clinical trial (NCT04518410), non-hospitalized participants with symptomatic SARS-CoV-2 infection were given bamlanivimab (700 mg or 7,000 mg) or placebo treatment. Here¸ we report that treatment-emergent resistance mutations [detected through targeted Spike (S) gene next-generation sequencing] were significantly more likely to be detected after bamlanivimab 700 mg treatment compared with the placebo group (7% of 111 vs 0% of 112 participants, P = 0.003). No treatment-emergent resistance mutations among the 48 participants who received 7,000 mg bamlanivimab were recorded. Participants in which emerging mAb resistant virus mutations were identified showed significantly higher pretreatment nasopharyngeal and anterior nasal viral loads. Daily respiratory tract viral sampling through study day 14 showed the dynamic nature of in vivo SARS-CoV-2 infection and indicated a rapid and sustained viral rebound after the emergence of resistance mutations. Participants with emerging bamlanivimab resistance often accumulated additional polymorphisms found in current variants of concern/interest that are associated with immune escape. These results highlight the potential for rapid emergence of resistance during mAb monotherapy treatment that results in prolonged high-level respiratory tract viral loads. Assessment of viral resistance should be prioritized during the development and clinical implementation of antiviral treatments for COVID-19.
Collapse
Affiliation(s)
- Manish C Choudhary
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kara W Chew
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Rinki Deo
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - James P Flynn
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - James Regan
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Charles R Crain
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Carlee Moser
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Justin Ritz
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ruy M Ribeiro
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Ruian Ke
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Joan A Dragavon
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | - Ajay Nirula
- Lilly Research Laboratories, San Diego, CA, USA
| | | | - Alexander L Greninger
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Courtney V Fletcher
- Center for Drug Discovery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eric S Daar
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - David A Wohl
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Joseph J Eron
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Judith S Currier
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Urvi M Parikh
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Scott F Sieg
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Alan S Perelson
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Robert W Coombs
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Davey M Smith
- Department of Medicine, University of California, San Diego, CA, USA.
| | - Jonathan Z Li
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Shrestha L, Lin MJ, Xie H, Mills MG, Mohamed Bakhash SA, Gaur VP, Livingston RJ, Castor J, Bruce EA, Botten JW, Huang ML, Jerome KR, Greninger AL, Roychoudhury P. Clinical Performance Characteristics of the Swift Normalase Amplicon Panel for Sensitive Recovery of Severe Acute Respiratory Syndrome Coronavirus 2 Genomes. J Mol Diagn 2022; 24:963-976. [PMID: 35863699 PMCID: PMC9290336 DOI: 10.1016/j.jmoldx.2022.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/24/2022] [Accepted: 05/27/2022] [Indexed: 11/18/2022] Open
Abstract
Amplicon-based sequencing methods are central in characterizing the diversity, transmission, and evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but need to be rigorously assessed for clinical utility. Herein, we validated the Swift Biosciences' SARS-CoV-2 Swift Normalase Amplicon Panels using remnant clinical specimens. High-quality genomes meeting our established library and sequence quality criteria were recovered from positive specimens, with 95% limit of detection of 40.08 SARS-CoV-2 copies/PCR. Breadth of genome recovery was evaluated across a range of CT values (11.3 to 36.7; median, 21.6). Of 428 positive samples, 413 (96.5%) generated genomes with <10% unknown bases, with a mean genome coverage of 13,545× ± SD 8382×. No genomes were recovered from PCR-negative specimens (n = 30) or from specimens positive for non-SARS-CoV-2 respiratory viruses (n = 20). Compared with whole-genome shotgun metagenomic sequencing (n = 14) or Sanger sequencing for the spike gene (n = 11), pairwise identity between consensus sequences was 100% in all cases, with highly concordant allele frequencies (R2 = 0.99) between Swift and shotgun libraries. When samples from different clades were mixed at varying ratios, expected variants were detected even in 1:99 mixtures. When deployed as a clinical test, 268 tests were performed in the first 23 weeks, with a median turnaround time of 11 days, ordered primarily for outbreak investigations and infection control.
Collapse
Affiliation(s)
- Lasata Shrestha
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Michelle J Lin
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Hong Xie
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Margaret G Mills
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Shah A Mohamed Bakhash
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Vinod P Gaur
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Robert J Livingston
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Jared Castor
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Emily A Bruce
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, Vermont
| | - Jason W Botten
- Department of Medicine, University of Vermont, Burlington, Vermont
| | - Meei-Li Huang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Keith R Jerome
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Alexander L Greninger
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| | - Pavitra Roychoudhury
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| |
Collapse
|
15
|
Chew KW, Moser C, Daar ES, Wohl DA, Li JZ, Coombs RW, Ritz J, Giganti M, Javan AC, Li Y, Choudhary MC, Deo R, Malvestutto C, Klekotka P, Price K, Nirula A, Fischer W, Bala V, Ribeiro RM, Perelson AS, Fletcher CV, Eron JJ, Currier JS, Hughes MD, Smith DM. Antiviral and clinical activity of bamlanivimab in a randomized trial of non-hospitalized adults with COVID-19. Nat Commun 2022; 13:4931. [PMID: 35995785 PMCID: PMC9395368 DOI: 10.1038/s41467-022-32551-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/04/2022] [Indexed: 01/25/2023] Open
Abstract
Anti-SARS-CoV-2 monoclonal antibodies are mainstay COVID-19 therapeutics. Safety, antiviral, and clinical efficacy of bamlanivimab were evaluated in the randomized controlled trial ACTIV-2/A5401. Non-hospitalized adults were randomized 1:1 within 10 days of COVID-19 symptoms to bamlanivimab or blinded-placebo in two dose-cohorts (7000 mg, n = 94; 700 mg, n = 223). No differences in bamlanivimab vs placebo were observed in the primary outcomes: proportion with undetectable nasopharyngeal SARS-CoV-2 RNA at days 3, 7, 14, 21, and 28 (risk ratio = 0.82-1.05 for 7000 mg [p(overall) = 0.88] and 0.81-1.21 for 700 mg [p(overall) = 0.49]), time to symptom improvement (median 21 vs 18.5 days [p = 0.97], 7000 mg; 24 vs 20.5 days [p = 0.08], 700 mg), or grade 3+ adverse events. However, bamlanivimab was associated with lower day 3 nasopharyngeal viral levels and faster reductions in inflammatory markers and viral decay by modeling. This study provides evidence of faster reductions in nasopharyngeal SARS-CoV-2 RNA levels but not shorter symptom durations in non-hospitalized adults with early variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Kara W Chew
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, USA.
| | - Carlee Moser
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eric S Daar
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - David A Wohl
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Jonathan Z Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert W Coombs
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Justin Ritz
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mark Giganti
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Yijia Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Manish C Choudhary
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rinki Deo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - William Fischer
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Veenu Bala
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, NE, USA
- Clinical Pharmacology & Pharmacometrics, Jounce Therapeutics, Cambridge, MA, USA
| | - Ruy M Ribeiro
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Alan S Perelson
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Courtney V Fletcher
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Joseph J Eron
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Judith S Currier
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Davey M Smith
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
16
|
Deo R, Choudhary MC, Moser C, Ritz J, Daar ES, Wohl DA, Greninger AL, Eron JJ, Currier JS, Hughes MD, Smith DM, Chew KW, Li JZ. Viral and Symptom Rebound in Untreated COVID-19 Infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.08.01.22278278. [PMID: 35982660 PMCID: PMC9387151 DOI: 10.1101/2022.08.01.22278278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background There are reports of viral RNA and symptom rebound in people with COVID-19 treated with nirmatrelvir/ritonavir. Since the natural course of viral and symptom trajectories of COVID-19 has not been well described, we evaluated the incidence of viral and symptom rebound in untreated outpatients with mild-moderate COVID-19. Methods The study population included 568 participants enrolled in the ACTIV-2/A5401 platform trial who received placebo. Anterior nasal swabs were collected for SARS-CoV-2 RNA testing on days 0-14, 21 and 28. Participants recorded the severity of 13 targeted symptoms daily from day 0 to 28. Viral rebound was defined as ≥0.5 log10 viral RNA copies/mL increase and symptom rebound was defined as a 4-point total symptom score increase from baseline. Baseline was defined as study day 4 (primary analysis) or 8 days from symptom onset (secondary analysis). Findings In both the primary and secondary analyses, 12% of participants had viral rebound. Viral rebounders were older than non-rebounders (median 54 vs 47 years, P=0.04). Symptom rebound occurred in 27% of participants after initial symptom improvement and in 10% of participants after initial symptom resolution. The combination of high-level viral rebound to ≥5.0 log10 RNA copies/mL and symptom rebound after initial improvement was observed in 1-2% of participants. Interpretation Viral RNA rebound or symptom relapse in the absence of antiviral treatment is common, but the combination of high-level viral and symptom rebound is rare.
Collapse
Affiliation(s)
- Rinki Deo
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Carlee Moser
- Harvard T.H. Chan School of Public Health, Boston, MA
| | - Justin Ritz
- Harvard T.H. Chan School of Public Health, Boston, MA
| | - Eric S Daar
- Lundquist Institute at Harbor-University of California, Los Angeles Medical Center, Torrance, CA
| | | | | | | | - Judith S Currier
- David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | | | | | - Kara W Chew
- David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | - Jonathan Z Li
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
17
|
Penney J, Jung A, Koethe B, Doron S. Cycle threshold values and SARS-CoV-2: Relationship to demographic characteristics and disease severity. J Med Virol 2022; 94:3978-3981. [PMID: 35355294 PMCID: PMC9088631 DOI: 10.1002/jmv.27752] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/01/2022] [Accepted: 03/29/2022] [Indexed: 12/15/2022]
Abstract
Cycle threshold (Ct), or the number of cycles required to amplify viral RNA to a detectable level, provides an estimate of viral load. Previous studies have demonstrated mixed results in regard to the association between SARS-CoV-2 Ct from real-time reverse transcriptase PCR (rRT-PCR) testing to patient outcomes, and there is less data on the relationship between Ct and patient characteristics. This was a retrospective study of 256 patients tested at a tertiary care emergency department from March to July 2020 via nasopharyngeal rRT-PCR testing utilizing the Abbott M2000 SARS-CoV-2 assay. Kruskal-Wallis, univariable, and multivariable logistic regression were used where appropriate for analysis. There were no significant differences in Ct value by demographic characteristics including age, sex, race, or ethnicity. Ct increased with time since symptom onset (p < 0.001), and increasing Ct was associated with increased odds of severe disease (odds ratio: 1.05, 95% confidence interval: 1.0-1.11). Ct was not found to be associated with patient demographic characteristics and increasing Ct was found to be associated with increased odds of severe disease. Continued study will allow us to better comprehend the complex factors that contribute to the risk for severe outcomes due to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jessica Penney
- Division of Geographic Medicine and Infectious DiseaseTufts Medical CenterBostonMassachusettsUSA
| | - Amanda Jung
- Tufts School of MedicineTufts University School of Public HealthBostonMassachusettsUSA
| | - Benjamin Koethe
- Biostatistics, Epidemiology and Research Design Center, Tufts Clinical and Translational Science InstituteBostonMassachusettsUSA
| | - Shira Doron
- Division of Geographic Medicine and Infectious DiseaseTufts Medical CenterBostonMassachusettsUSA
| |
Collapse
|
18
|
Abstract
Scaling up SARS-CoV-2 testing during the COVID-19 pandemic was critical to maintaining clinical operations and an open society. Pooled testing and automation were two critical strategies used by laboratories to meet the unprecedented demand. Here, we review these and other cutting-edge strategies that sought to expand SARS-CoV-2 testing capacity while maintaining high individual test performance.
Collapse
Affiliation(s)
- Sanchita Das
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Karen M Frank
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA.
| |
Collapse
|
19
|
Utility of the cycle threshold in anticipating the next phase of the coronavirus disease 2019 (COVID-19) pandemic. Infect Control Hosp Epidemiol 2022; 43:800-801. [DOI: 10.1017/ice.2022.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
20
|
Campbell MR, Binnicker MJ. Analytic and clinical performance of major commercial SARS-CoV-2 molecular assays in the United States. Clin Lab Med 2022; 42:129-145. [PMID: 35636818 PMCID: PMC8858713 DOI: 10.1016/j.cll.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
21
|
Clinical evaluation of the automated Abbott RealTime SARS-CoV-2, Alinity m SARS-CoV-2, and Alinity m Resp-4-Plex assays. J Virol Methods 2022; 299:114338. [PMID: 34695479 PMCID: PMC8532389 DOI: 10.1016/j.jviromet.2021.114338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/15/2021] [Accepted: 10/15/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Detection of SARS-CoV-2 infections relies on the use of sensitive, accurate and high throughput RT-PCR assays. OBJECTIVES We assessed the analytical performance of the Abbott RealTime SARS-CoV-2 (RT-SARS), Alinity m SARS-CoV-2 (AlinSARS) assays and compared the clinical performance of the RT-SARS, AlinSARS, and Alinity m Resp-4-Plex (Alin4Plex) assays to the Seegene Allplex assay (Allplex) and an inhouse test (Inhouse). RESULTS We found 100 % positive percent agreement (PPA) and 100 % negative percent agreement (NPA) comparing RT-SARS and Allplex. RT-SARS, AlinSARS and Inhouse showed 100 % NPA and 100 % PPA across all assays, except for the RdRp target of Inhouse (PPA = 84 %). Similarly, Alin4Plex and Allplex showed high agreement with specimens containing either SARS-CoV-2, influenza A, influenza B, or RSV. Detection rates of 100 % for SARS-CoV-2 at 50 copies/mL, high precision, and no cross-reactivity with non-SARS-CoV-2 respiratory pathogens were observed for RT-SARS and AlinSARS. AlinSARS detected SARS-CoV-2 in spiked throat washes and in specimens infected with SARS-CoV-2 Alpha or Beta variants. CONCLUSIONS The newly developed RT-SARS, AlinSARS, and Alin4Plex assays proved to be useful for detecting SARS-CoV-2 RNA in clinical samples.
Collapse
|
22
|
Orf GS, Forberg K, Meyer TV, Mowerman I, Mohaimani A, Faron ML, Jennings C, Landay AL, Goldstein DY, Fox AS, Berg MG, Cloherty GA. SNP and Phylogenetic Characterization of Low Viral Load SARS-CoV-2 Specimens by Target Enrichment. FRONTIERS IN VIROLOGY 2021. [DOI: 10.3389/fviro.2021.765974] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background: Surveillance of SARS-CoV-2 across the globe has enabled detection of new variants and informed the public health response. With highly sensitive methods like qPCR widely adopted for diagnosis, the ability to sequence and characterize specimens with low titers needs to keep pace.Methods: Nucleic acids extracted from nasopharyngeal swabs collected from four sites in the United States in early 2020 were converted to NGS libraries to sequence SARS-CoV-2 genomes using metagenomic and xGen target enrichment approaches. Single nucleotide polymorphism (SNP) analysis and phylogeny were used to determine clade assignments and geographic origins of strains.Results: SARS-CoV-2-specific xGen enrichment enabled full genome coverage for 87 specimens with Ct values <29, corresponding to viral loads of >10,000 cp/ml. For samples with viral loads between 103 and 106 cp/ml, the median genome coverage for xGen was 99.1%, sequence depth was 605X, and the “on-target” rate was 57 ± 21%, compared to 13%, 2X and 0.001 ± 0.016%, respectively, for metagenomic sequencing alone. Phylogenetic analysis revealed the presence of most clades that existed at the time of the study, though clade GH dominated in the Midwest.Conclusions: Even as vaccines are being widely distributed, a high case load of SARS-CoV-2 infection persists around the world. Viral genetic surveillance has succeeded in warning the public of new variants in circulation and ensured that diagnostic tools remain resilient to a steadily increasing number of mutations. Target capture offers a means of characterizing low viral load samples which would normally pose a challenge for metagenomic sequencing.
Collapse
|
23
|
Gohil SK, Quan KA, Madey KM, King-Adelsohn S, Tjoa T, Tifrea D, Crews BO, Monuki ES, Khan S, Schubl SD, Bittencourt CE, Detweiler N, Chang W, Willis L, Khusbu U, Saturno A, Rezk SA, Figueroa C, Jain A, Assis R, Felgner P, Edwards R, Hsieh L, Forthal D, Wilson WC, Stamos MJ, Huang SS. Infection prevention strategies are highly protective in COVID-19 units while main risks to healthcare professionals come from coworkers and the community. Antimicrob Resist Infect Control 2021; 10:163. [PMID: 34809702 PMCID: PMC8608236 DOI: 10.1186/s13756-021-01031-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/27/2021] [Indexed: 11/19/2022] Open
Abstract
Background Early evaluations of healthcare professional (HCP) COVID-19 risk occurred during insufficient personal protective equipment and disproportionate testing, contributing to perceptions of high patient-care related HCP risk. We evaluated HCP COVID-19 seropositivity after accounting for community factors and coworker outbreaks. Methods Prior to universal masking, we conducted a single-center retrospective cohort plus cross-sectional study. All HCP (1) seen by Occupational Health for COVID-like symptoms (regardless of test result) or assigned to (2) dedicated COVID-19 units, (3) units with a COVID-19 HCP outbreak, or (4) control units from 01/01/2020 to 04/15/2020 were offered serologic testing by an FDA-authorized assay plus a research assay against 67 respiratory viruses, including 11 SARS-CoV-2 antigens. Multivariable models assessed the association of demographics, job role, comorbidities, care of a COVID-19 patient, and geocoded socioeconomic status with positive serology. Results Of 654 participants, 87 (13.3%) were seropositive; among these 60.8% (N = 52) had never cared for a COVID-19 patient. Being male (OR 1.79, CI 1.05–3.04, p = 0.03), working in a unit with a HCP-outbreak unit (OR 2.21, CI 1.28–3.81, p < 0.01), living in a community with low owner-occupied housing (OR = 1.63, CI = 1.00–2.64, p = 0.05), and ethnically Latino (OR 2.10, CI 1.12–3.96, p = 0.02) were positively-associated with COVID-19 seropositivity, while working in dedicated COVID-19 units was negatively-associated (OR 0.53, CI = 0.30–0.94, p = 0.03). The research assay identified 25 additional seropositive individuals (78 [12%] vs. 53 [8%], p < 0.01). Conclusions Prior to universal masking, HCP COVID-19 risk was dominated by workplace and community exposures while working in a dedicated COVID-19 unit was protective, suggesting that infection prevention protocols prevent patient-to-HCP transmission. Article summary Prior to universal masking, HCP COVID-19 risk was dominated by workplace and community exposures while working in a dedicated COVID-19 unit was protective, suggesting that infection prevention protocols prevent patient-to-HCP transmission. Supplementary Information The online version contains supplementary material available at 10.1186/s13756-021-01031-5.
Collapse
Affiliation(s)
- Shruti K Gohil
- Epidemiology and Infection Prevention Program, Irvine Health (UC Irvine Health), University of California, Irvine, USA. .,Division of Infectious Diseases, Department of Medicine, Irvine School of Medicine, University of California, 100 Theory, Suite 120, Irvine, CA, 92617, USA.
| | - Kathleen A Quan
- Epidemiology and Infection Prevention Program, Irvine Health (UC Irvine Health), University of California, Irvine, USA
| | - Keith M Madey
- Epidemiology and Infection Prevention Program, Irvine Health (UC Irvine Health), University of California, Irvine, USA
| | | | - Tom Tjoa
- Division of Infectious Diseases, Department of Medicine, Irvine School of Medicine, University of California, 100 Theory, Suite 120, Irvine, CA, 92617, USA
| | - Delia Tifrea
- Pathology and Laboratory Medicine, UCI Health, Newport Beach, USA
| | - Bridgit O Crews
- Pathology and Laboratory Medicine, UCI Health, Newport Beach, USA
| | - Edwin S Monuki
- Pathology and Laboratory Medicine, UCI Health, Newport Beach, USA
| | - Saahir Khan
- Division of Infectious Diseases, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Sebastian D Schubl
- Department of Surgery, Irvine School of Medicine, University of California, Irvine, USA
| | | | - Neil Detweiler
- Pathology and Laboratory Medicine, UCI Health, Newport Beach, USA
| | - Wayne Chang
- Division of Occupational and Environmental Medicine, Irvine School of Medicine, University of California, Irvine, USA
| | - Lynn Willis
- Epidemiology and Infection Prevention Program, Irvine Health (UC Irvine Health), University of California, Irvine, USA
| | - Usme Khusbu
- Epidemiology and Infection Prevention Program, Irvine Health (UC Irvine Health), University of California, Irvine, USA
| | - Antonella Saturno
- Epidemiology and Infection Prevention Program, Irvine Health (UC Irvine Health), University of California, Irvine, USA
| | - Sherif A Rezk
- Pathology and Laboratory Medicine, UCI Health, Newport Beach, USA
| | - Cesar Figueroa
- Department of Surgery, Irvine School of Medicine, University of California, Irvine, USA
| | - Aarti Jain
- Department of Physiology and Biophysics, University of California, Irvine, USA
| | - Rafael Assis
- Department of Physiology and Biophysics, University of California, Irvine, USA
| | - Philip Felgner
- Department of Physiology and Biophysics, University of California, Irvine, USA
| | - Robert Edwards
- Pathology and Laboratory Medicine, UCI Health, Newport Beach, USA
| | - Lanny Hsieh
- Division of Infectious Diseases, Department of Medicine, Irvine School of Medicine, University of California, 100 Theory, Suite 120, Irvine, CA, 92617, USA
| | - Donald Forthal
- Division of Infectious Diseases, Department of Medicine, Irvine School of Medicine, University of California, 100 Theory, Suite 120, Irvine, CA, 92617, USA
| | | | - Michael J Stamos
- Department of Surgery, Irvine School of Medicine, University of California, Irvine, USA
| | - Susan S Huang
- Epidemiology and Infection Prevention Program, Irvine Health (UC Irvine Health), University of California, Irvine, USA.,Division of Infectious Diseases, Department of Medicine, Irvine School of Medicine, University of California, 100 Theory, Suite 120, Irvine, CA, 92617, USA
| |
Collapse
|
24
|
Grau S, Ferrández O, Echeverría-Esnal D, Maldonado R, Puig B, Ramirez A, Canal M, Montero A, González C, Herranz M, Masclans JR, Horcajada JP, Padilla E. SARS-CoV-2 could be spread through hospital medication dispensed to patients: A prospective observational study. Medicine (Baltimore) 2021; 100:e27592. [PMID: 34766561 PMCID: PMC8589228 DOI: 10.1097/md.0000000000027592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 08/02/2021] [Accepted: 10/08/2021] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT Our objective was to analyze in vitro the persistence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in the packaging material of the drugs dispensed to hospital wards. Additionally, to evaluate if the protection with a double plastic bag prevents the contamination of the medication dispensed to an intensive care unit (ICU).On the first part, different materials containing different drugs within an ICU were sampled to confirm the lack of contamination by SARS-CoV-2. The confirmation of the virus was performed using real time reverse transcription polymerase chain reaction. As a control group, in the microbiology laboratory we inoculated the virus into the different surfaces containing the same drugs included in the first part. Samples were obtained with a sterile swab at 3, 6, 8, 10, 14, 21, and 30 days after inoculation and analyzed through real time reverse transcription polymerase chain reaction.None of the studied materials containing the drugs within an ICU was contaminated by SARS-CoV-2. In the second part, SARS-CoV-2 was found in all surfaces for up to 30 days.The use of double-bag unit-dose system to deliver medication in a pandemic seems effective to prevent the potential transmission of SARS-CoV-2. A striking SARS-CoV-2 RNA stability of up to 30 days was found in the surfaces containing the drugs.
Collapse
Affiliation(s)
- Santiago Grau
- Pharmacy Department, Hospital del Mar, Barcelona, Spain
- Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Infectious Pathology and Antimicrobials Research Group (IPAR), Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Spain
| | - Olivia Ferrández
- Pharmacy Department, Hospital del Mar, Barcelona, Spain
- Infectious Pathology and Antimicrobials Research Group (IPAR), Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Spain
| | - Daniel Echeverría-Esnal
- Pharmacy Department, Hospital del Mar, Barcelona, Spain
- Infectious Pathology and Antimicrobials Research Group (IPAR), Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, PRBB, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), PRBB, Barcelona, Spain
| | - Berta Puig
- Microbiology Department, Laboratori de Referència de Catalunya, Barcelona, Spain
| | - Aida Ramirez
- Microbiology Department, Laboratori de Referència de Catalunya, Barcelona, Spain
| | - Mireia Canal
- Microbiology Department, Laboratori de Referència de Catalunya, Barcelona, Spain
| | | | - Cristina González
- Epidemiology and Evaluation Department, Parc de Salut Mar, Barcelona, Spain
| | - Milagros Herranz
- Epidemiology and Evaluation Department, Parc de Salut Mar, Barcelona, Spain
| | - Joan Ramon Masclans
- Critical Care Department, Hospital del Mar, Barcelona, Spain
- Critical Ill Patient Research Group (GREPAC), Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Universitat Autònoma de Barcelona (UAB), Spain
| | - Juan Pablo Horcajada
- Infectious Pathology and Antimicrobials Research Group (IPAR), Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Spain
- Infectious Diseases Deparment, Hospital del Mar, Universitat Autònoma de Barcelona, Passeig Marítim 25, Barcelona, Spain
| | - Eduardo Padilla
- Microbiology Department, Laboratori de Referència de Catalunya, Barcelona, Spain
| |
Collapse
|
25
|
Berg MG, Zhen W, Lucic D, Degli-Angeli EJ, Anderson M, Forberg K, Olivo A, Sheikh F, Toolsie D, Greninger AL, Cloherty GA, Coombs RW, Berry GJ. Development of the RealTime SARS-CoV-2 quantitative Laboratory Developed Test and correlation with viral culture as a measure of infectivity. J Clin Virol 2021; 143:104945. [PMID: 34450558 PMCID: PMC8367731 DOI: 10.1016/j.jcv.2021.104945] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/21/2021] [Accepted: 08/09/2021] [Indexed: 12/23/2022]
Abstract
While diagnosis of COVID-19 relies on qualitative molecular testing for the absence or presence of SARS-CoV-2 RNA, quantitative viral load determination for SARS-CoV-2 has many potential applications in antiviral therapy and vaccine trials as well as implications for public health and quarantine guidance. To date, no quantitative SARS-CoV-2 viral load tests have been authorized for clinical use by the FDA. In this study, we modified the FDA emergency use authorized qualitative RealTime SARS-CoV-2 assay into a quantitative SARS-CoV-2 Laboratory Developed Test (LDT) using newly developed Abbott SARS-CoV-2 calibration standards. Both analytical and clinical performance of this SARS-CoV-2 quantitative LDT was evaluated using nasopharyngeal swabs (NPS). We further assessed the correlation between Ct and the ability to culture virus on Vero CCL81 cells. The SARS-CoV-2 quantitative LDT demonstrated high linearity with R2 value of 0.992, high inter- and intra-assay reproducibility across the dynamic range (SDs ± 0.08-0.14 log10 copies/mL for inter-assay reproducibility and ± 0.09 to 0.19 log10 copies/mL for intra-assay reproducibility). Lower limit of detection was determined as 1.90 log10 copies/mL. The highest Ct at which CPE was detected ranged between 28.21-28.49, corresponding to approximately 4.2 log10 copies/mL. Quantitative tests, validated against viral culture capacity, may allow more accurate identification of individuals with and without infectious viral shedding from the respiratory tract.
Collapse
Affiliation(s)
| | - Wei Zhen
- Northwell Health Laboratories, Lake Success, NY, United States
| | | | - Emily J Degli-Angeli
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, WA, United States
| | | | - Kenn Forberg
- Abbott Laboratories, Abbott Park, IL, United States
| | - Ana Olivo
- Abbott Laboratories, Abbott Park, IL, United States
| | - Farah Sheikh
- Northwell Health Laboratories, Lake Success, NY, United States
| | - Dan Toolsie
- Abbott Molecular, Des Plaines, IL, United States
| | - Alexander L Greninger
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, WA, United States
| | | | - Robert W Coombs
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, WA, United States
| | - Gregory J Berry
- Northwell Health Laboratories, Lake Success, NY, United States
| |
Collapse
|
26
|
Choudhary MC, Chew KW, Deo R, Flynn JP, Regan J, Crain CR, Moser C, Hughes M, Ritz J, Ribeiro RM, Ke R, Dragavon JA, Javan AC, Nirula A, Klekotka P, Greninger AL, Fletcher CV, Daar ES, Wohl DA, Eron JJ, Currier JS, Parikh UM, Sieg SF, Perelson AS, Coombs RW, Smith DM, Li JZ. Emergence of SARS-CoV-2 Resistance with Monoclonal Antibody Therapy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.09.03.21263105. [PMID: 34545376 PMCID: PMC8452115 DOI: 10.1101/2021.09.03.21263105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Resistance mutations to monoclonal antibody (mAb) therapy has been reported, but in the non-immunosuppressed population, it is unclear if in vivo emergence of SARS-CoV-2 resistance mutations alters either viral replication dynamics or therapeutic efficacy. In ACTIV-2/A5401, non-hospitalized participants with symptomatic SARS-CoV-2 infection were randomized to bamlanivimab (700mg or 7000mg) or placebo. Treatment-emergent resistance mutations were significantly more likely detected after bamlanivimab 700mg treatment than placebo (7% of 111 vs 0% of 112 participants, P=0.003). There were no treatment-emergent resistance mutations among the 48 participants who received bamlanivimab 7000mg. Participants with emerging mAb resistant virus had significantly higher pre-treatment nasopharyngeal and anterior nasal viral load. Intensive respiratory tract viral sampling revealed the dynamic nature of SARS-CoV-2 evolution, with evidence of rapid and sustained viral rebound after emergence of resistance mutations, and worsened symptom severity. Participants with emerging bamlanivimab resistance often accumulated additional polymorphisms found in current variants of concern/interest and associated with immune escape. These results highlight the potential for rapid emergence of resistance during mAb monotherapy treatment, resulting in prolonged high level respiratory tract viral loads and clinical worsening. Careful virologic assessment should be prioritized during the development and clinical implementation of antiviral treatments for COVID-19.
Collapse
|
27
|
Kanwar N, Banerjee D, Sasidharan A, Abdulhamid A, Larson M, Lee B, Selvarangan R, Liesman RM. Comparison of diagnostic performance of five molecular assays for detection of SARS-CoV-2. Diagn Microbiol Infect Dis 2021; 101:115518. [PMID: 34481324 PMCID: PMC8343369 DOI: 10.1016/j.diagmicrobio.2021.115518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/26/2021] [Accepted: 08/01/2021] [Indexed: 12/01/2022]
Abstract
We compared the performance of the Abbott Real Time SARS-CoV-2 assay (Abbott assay), Aptima™ SARS-CoV-2 assay (Aptima assay), BGI Real-Time SARS-CoV-2 assay (BGI assay), Lyra® SARS-CoV-2 assay (Lyra assay), and DiaSorin Simplexa™ COVID assay for SARS-CoV-2 detection. Residual nasopharyngeal samples (n = 201) submitted for routine SARS-CoV-2 testing by Simplexa assay during June-July 2020 and January 2021 were salvaged. Aliquots were tested on other assays and compared against the CDC 2019-nCoV Real-Time RT-PCR assay. Viral load in positive samples was determined by droplet digital PCR. Among 201 samples, 99 were positive and 102 were negative by the CDC assay. The Aptima and Abbott assays exhibited the highest positive percent agreement (PPA) at 98.9% while the BGI assay demonstrated the lowest PPA of 89.9% with 10 missed detections. Negative percent agreement for all 5 platforms was comparable, ranging from 96.1% to 100%. The performance of all five assays was comparable.
Collapse
Affiliation(s)
- Neena Kanwar
- Department of Pathology and Laboratory Medicine, Children's Mercy, Kansas City, MO, USA; School of Medicine, University of Missouri- Kansas City, MO, USA
| | - Dithi Banerjee
- Department of Pathology and Laboratory Medicine, Children's Mercy, Kansas City, MO, USA; School of Medicine, University of Missouri- Kansas City, MO, USA
| | - Anjana Sasidharan
- Department of Pathology and Laboratory Medicine, Children's Mercy, Kansas City, MO, USA
| | - Ayah Abdulhamid
- Department of Pathology and Laboratory Medicine, Children's Mercy, Kansas City, MO, USA
| | - Marissa Larson
- Department of Pathology and Laboratory Medicine, University of Kansas Health System, Kansas City, KS, USA
| | - Brian Lee
- School of Medicine, University of Missouri- Kansas City, MO, USA; Division of Health Services and Outcomes Research, Children's Mercy, Kansas City, MO, USA
| | - Rangaraj Selvarangan
- Department of Pathology and Laboratory Medicine, Children's Mercy, Kansas City, MO, USA; School of Medicine, University of Missouri- Kansas City, MO, USA; Department of Pathology and Laboratory Medicine, University of Kansas Health System, Kansas City, KS, USA.
| | - Rachael M Liesman
- Department of Pathology and Laboratory Medicine, University of Kansas Health System, Kansas City, KS, USA.
| |
Collapse
|
28
|
Callahan C, Lee RA, Lee GR, Zulauf K, Kirby JE, Arnaout R. Nasal Swab Performance by Collection Timing, Procedure, and Method of Transport for Patients with SARS-CoV-2. J Clin Microbiol 2021; 59:e0056921. [PMID: 34076471 PMCID: PMC8373031 DOI: 10.1128/jcm.00569-21] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/30/2021] [Indexed: 12/23/2022] Open
Abstract
The urgent need for large-scale diagnostic testing for SARS-CoV-2 has prompted interest in sample collection methods of sufficient sensitivity to replace nasopharynx (NP) sampling. Nasal swab samples are an attractive alternative; however, previous studies have disagreed over how nasal sampling performs relative to NP sampling. Here, we compared nasal versus NP specimens collected by health care workers in a cohort of individuals clinically suspected of COVID-19 as well as SARS-CoV-2 reverse transcription (RT)-PCR-positive outpatients undergoing follow-up. We compared subjects being seen for initial evaluation versus follow-up, two different nasal swab collection protocols, and three different transport conditions, including traditional viral transport media (VTM) and dry swabs, on 307 total study participants. We compared categorical results and viral loads to those from standard NP swabs collected at the same time from the same patients. All testing was performed by RT-PCR on the Abbott SARS-CoV-2 RealTime emergency use authorization (EUA) (limit of detection [LoD], 100 copies viral genomic RNA/ml transport medium). We found low concordance overall, with Cohen's kappa (κ) of 0.49, with high concordance only for subjects with very high viral loads. We found medium concordance for testing at initial presentation (κ = 0.68) and very low concordance for follow-up testing (κ = 0.27). Finally, we show that previous reports of high concordance may have resulted from measurement using assays with sensitivity of ≥1,000 copies/ml. These findings suggest nasal-swab testing be used for situations in which viral load is expected to be high, as we demonstrate that nasal swab testing is likely to miss patients with low viral loads.
Collapse
Affiliation(s)
- Cody Callahan
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Rose A. Lee
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Division of Infectious Diseases, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ghee Rye Lee
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Kate Zulauf
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - James E. Kirby
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ramy Arnaout
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Clinical Informatics, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Peng B, Zhou W, Pettit RW, Yu P, Matos PG, Greninger AL, McCashin J, Amos CI. Reducing COVID-19 quarantine with SARS-CoV-2 testing: a simulation study. BMJ Open 2021; 11:e050473. [PMID: 34272225 PMCID: PMC8290949 DOI: 10.1136/bmjopen-2021-050473] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/29/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To evaluate the effectiveness of SARS-CoV-2 testing on shortening the duration of quarantines for COVID-19 and to identify the most effective choices of testing schedules. DESIGN We performed extensive simulations to evaluate the performance of quarantine strategies when one or more SARS-CoV-2 tests were administered during the quarantine. Simulations were based on statistical models for the transmissibility and viral loads of SARS-CoV-2 infections and the sensitivities of available testing methods. Sensitivity analyses were performed to evaluate the impact of perturbations in model assumptions on the outcomes of optimal strategies. RESULTS We found that SARS-CoV-2 testing can effectively reduce the length of a quarantine without compromising safety. A single reverse transcription-PCR (RT-PCR) test performed before the end of quarantine can reduce quarantine duration to 10 days. Two tests can reduce the duration to 8 days, and three highly sensitive RT-PCR tests can justify a 6-day quarantine. More strategic testing schedules and longer quarantines are needed if tests are administered with less-sensitive RT-PCR tests or antigen tests. Shorter quarantines can be used for applications that tolerate a residual postquarantine transmission risk comparable to a 10-day quarantine. CONCLUSIONS Testing could substantially reduce the length of isolation, reducing the physical and mental stress caused by lengthy quarantines. With increasing capacity and lowered costs of SARS-CoV-2 tests, test-assisted quarantines could be safer and more cost-effective than 14-day quarantines and warrant more widespread use.
Collapse
Affiliation(s)
- Bo Peng
- Institute for Clinical & Translational Research, Baylor College of Medicine, Houston, Texas, USA
| | - Wen Zhou
- Institute for Clinical & Translational Research, Baylor College of Medicine, Houston, Texas, USA
| | - Rowland W Pettit
- Institute for Clinical & Translational Research, Baylor College of Medicine, Houston, Texas, USA
| | - Patrick Yu
- Corporate Medical Advisors, Houston, Texas, USA
| | | | - Alexander L Greninger
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | | | - Christopher I Amos
- Institute for Clinical & Translational Research, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
30
|
Safiabadi Tali SH, LeBlanc JJ, Sadiq Z, Oyewunmi OD, Camargo C, Nikpour B, Armanfard N, Sagan SM, Jahanshahi-Anbuhi S. Tools and Techniques for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2)/COVID-19 Detection. Clin Microbiol Rev 2021; 34:e00228-20. [PMID: 33980687 PMCID: PMC8142517 DOI: 10.1128/cmr.00228-20] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory disease coronavirus 2 (SARS-CoV-2), has led to millions of confirmed cases and deaths worldwide. Efficient diagnostic tools are in high demand, as rapid and large-scale testing plays a pivotal role in patient management and decelerating disease spread. This paper reviews current technologies used to detect SARS-CoV-2 in clinical laboratories as well as advances made for molecular, antigen-based, and immunological point-of-care testing, including recent developments in sensor and biosensor devices. The importance of the timing and type of specimen collection is discussed, along with factors such as disease prevalence, setting, and methods. Details of the mechanisms of action of the various methodologies are presented, along with their application span and known performance characteristics. Diagnostic imaging techniques and biomarkers are also covered, with an emphasis on their use for assessing COVID-19 or monitoring disease severity or complications. While the SARS-CoV-2 literature is rapidly evolving, this review highlights topics of interest that have occurred during the pandemic and the lessons learned throughout. Exploring a broad armamentarium of techniques for detecting SARS-CoV-2 will ensure continued diagnostic support for clinicians, public health, and infection prevention and control for this pandemic and provide advice for future pandemic preparedness.
Collapse
Affiliation(s)
- Seyed Hamid Safiabadi Tali
- Department of Chemical and Materials Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
- Department of Mechanical, Industrial, and Aerospace Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
| | - Jason J LeBlanc
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medicine (Infectious Diseases), Dalhousie University, Halifax, Nova Scotia, Canada
- Division of Microbiology, Department of Pathology and Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
| | - Zubi Sadiq
- Department of Chemical and Materials Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
| | - Oyejide Damilola Oyewunmi
- Department of Chemical and Materials Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
| | - Carolina Camargo
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | - Bahareh Nikpour
- Department of Electrical and Computer Engineering, McGill University, Montréal, Québec, Canada
| | - Narges Armanfard
- Department of Electrical and Computer Engineering, McGill University, Montréal, Québec, Canada
- Mila-Quebec AI Institute, Montréal, Québec, Canada
| | - Selena M Sagan
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Sana Jahanshahi-Anbuhi
- Department of Chemical and Materials Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
| |
Collapse
|
31
|
Performance of three polymerase chain reaction-based assays for detection of SARS-CoV-2 in different upper respiratory tract specimens. Diagn Microbiol Infect Dis 2021; 101:115441. [PMID: 34186320 PMCID: PMC8160280 DOI: 10.1016/j.diagmicrobio.2021.115441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 12/04/2022]
Abstract
To meet the testing demands and overcome supply chain issues during the SARS-CoV-2 pandemic, many clinical laboratories validated multiple SARS-CoV-2 molecular testing platforms. Here, we compare three different molecular assays for SARS-CoV-2 that received emergency use authorization (EUA) from the U.S. Food and Drug Administration. In order to determine the agreement among Roche cobas® SARS-CoV-2 Test (Cobas), Abbott RealTime SARS-CoV-2 assay (ART), and Mayo Clinic Laboratory SARS-CoV-2 Molecular Detection Assay (Mayo LDT), 100 each of anterior nares (AN), nasopharyngeal (NP), oropharyngeal (OP), and NP+OP swabs were tested on each platform. The consensus result was defined as agreement by 2 or more methods. Furthermore, 30 positive NP swabs from each molecular platform (n = 90 total) were tested on the three platforms to determine the PPA among positive samples. ART platform called more specimens positive than the other two platforms. All three assays performed with greater than 90% agreement for NP specimens throughout the study.
Collapse
|
32
|
Mardian Y, Kosasih H, Karyana M, Neal A, Lau CY. Review of Current COVID-19 Diagnostics and Opportunities for Further Development. Front Med (Lausanne) 2021; 8:615099. [PMID: 34026773 PMCID: PMC8138031 DOI: 10.3389/fmed.2021.615099] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
Diagnostic testing plays a critical role in addressing the coronavirus disease 2019 (COVID-19) pandemic, caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Rapid and accurate diagnostic tests are imperative for identifying and managing infected individuals, contact tracing, epidemiologic characterization, and public health decision making. Laboratory testing may be performed based on symptomatic presentation or for screening of asymptomatic people. Confirmation of SARS-CoV-2 infection is typically by nucleic acid amplification tests (NAAT), which requires specialized equipment and training and may be particularly challenging in resource-limited settings. NAAT may give false-negative results due to timing of sample collection relative to infection, improper sampling of respiratory specimens, inadequate preservation of samples, and technical limitations; false-positives may occur due to technical errors, particularly contamination during the manual real-time polymerase chain reaction (RT-PCR) process. Thus, clinical presentation, contact history and contemporary phyloepidemiology must be considered when interpreting results. Several sample-to-answer platforms, including high-throughput systems and Point of Care (PoC) assays, have been developed to increase testing capacity and decrease technical errors. Alternatives to RT-PCR assay, such as other RNA detection methods and antigen tests may be appropriate for certain situations, such as resource-limited settings. While sequencing is important to monitor on-going evolution of the SARS-CoV-2 genome, antibody assays are useful for epidemiologic purposes. The ever-expanding assortment of tests, with varying clinical utility, performance requirements, and limitations, merits comparative evaluation. We herein provide a comprehensive review of currently available COVID-19 diagnostics, exploring their pros and cons as well as appropriate indications. Strategies to further optimize safety, speed, and ease of SARS-CoV-2 testing without compromising accuracy are suggested. Access to scalable diagnostic tools and continued technologic advances, including machine learning and smartphone integration, will facilitate control of the current pandemic as well as preparedness for the next one.
Collapse
Affiliation(s)
- Yan Mardian
- Indonesia Research Partnership on Infectious Disease (INA-RESPOND), Jakarta, Indonesia
| | - Herman Kosasih
- Indonesia Research Partnership on Infectious Disease (INA-RESPOND), Jakarta, Indonesia
| | - Muhammad Karyana
- Indonesia Research Partnership on Infectious Disease (INA-RESPOND), Jakarta, Indonesia
- National Institute of Health Research and Development, Ministry of Health, Republic of Indonesia, Jakarta, Indonesia
| | - Aaron Neal
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Chuen-Yen Lau
- National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
33
|
Heald-Sargent T, Muller WJ, Kociolek LK. Viral Loads of SARS-CoV-2 in Young Children-Reply. JAMA Pediatr 2021; 175:530-531. [PMID: 33523126 DOI: 10.1001/jamapediatrics.2020.5551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Taylor Heald-Sargent
- Division of Infectious Diseases, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois
| | - William J Muller
- Division of Infectious Diseases, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois.,Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Larry K Kociolek
- Division of Infectious Diseases, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois.,Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
34
|
Verification and Validation of SARS-CoV-2 Assay Performance on the Abbott m2000 and Alinity m Systems. J Clin Microbiol 2021; 59:JCM.03119-20. [PMID: 33568468 PMCID: PMC8091852 DOI: 10.1128/jcm.03119-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/09/2021] [Indexed: 12/19/2022] Open
Abstract
We verified the analytical performance of the Abbott RealTime SARS-CoV-2 assay on the m2000 system and compared its clinical performance to the CDC 2019-nCoV real-time PCR diagnostic panel and the Thermo Fisher TaqPath RT-PCR COVID-19 kit. We also performed a bridging study comparing the RealTime SARS-CoV-2 assay with the new Abbott Alinity m SARS-CoV-2 assay. A number of standards, reference materials, and commercially available controls were used for the analytical verification to confirm the limit of detection, linearity, and reproducibility. We used nasopharyngeal (NP) swab specimens collected in saline for the clinical verification and bridging studies. Overall, we found 91.2% positive percent agreement (PPA; 95% confidence interval [CI] = 76.2 to 98.14%) and a 100% negative percent agreement (NPA; 95% CI = 97.97 to 100%) between the results of the RealTime SARS-CoV-2 and CDC tests with 217 NP specimens (P = 0.13). We found a PPA of 100% (95% CI = 90.26 to 100%) and an NPA of 95.15% (95% CI = 83.47 to 99.4%) between the results of the RealTime and TaqPath tests with 77 NP specimens (P = 0.24). Finally, we tested 203 NP swab specimens for SARS-CoV-2 on the m2000 on the Alinity m systems. The PPA and NPA were 92.2% (95% CI = 85.3 to 96.59%) and 92% (95% CI = 84.8 to 96.5%), respectively (P = 0.4). Although cycle number (Cn) values obtained for the concordant positive samples were highly correlated (R 2 = 0.95), the Cn values were on average 14.14 higher on the Alinity m system due to the unread cycles with the RealTime SARS-CoV-2 assay.
Collapse
|
35
|
Johnson-Agbakwu CE, Eakin CM, Bailey CV, Sood S, Ali N, Doehrman P, Bhattarai B, Chambliss L, Coonrod DV. Severe acute respiratory syndrome coronavirus 2: a canary in the coal mine for public safety net hospitals. AJOG GLOBAL REPORTS 2021; 1:100009. [PMID: 33937884 PMCID: PMC7981272 DOI: 10.1016/j.xagr.2021.100009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/13/2021] [Accepted: 03/16/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The coronavirus disease 2019 pandemic has exposed disproportionate health inequities among underserved populations, including refugees. Public safety net healthcare systems play a critical role in facilitating access to care for refugees and informing coordinated public health prevention and mitigation efforts during a pandemic. OBJECTIVE This study aimed to evaluate the prevalence ratios of severe acute respiratory syndrome coronavirus 2 infection between refugee women and nonrefugee parturient patients admitted to the hospital for delivery. Here, we suspected that the burden of infection was disproportionately distributed across refugee communities that may act as sentinels for community outbreaks. STUDY DESIGN A cross-sectional study was conducted examining parturient women admitted to the maternity unit between May 6, 2020, and July 22, 2020, when universal testing for severe acute respiratory syndrome coronavirus 2 was first employed. Risk factors for severe acute respiratory syndrome 2 positivity were ascertained, disaggregated by refugee status, and other clinical and sociodemographic variables examined. Prevalence ratios were calculated and comparisons made to county-level community prevalence over the same period. RESULTS The positive test percentage at the county-level during this study period was 21.6%. Of 350 women admitted to the hospital for delivery, 33 (9.4%) tested positive for severe acute respiratory syndrome 2. When refugee status was determined, 45 women (12.8%) were identified as refugees. Of the 45 refugee women, 8 (17.8%) tested positive for severe acute respiratory syndrome 2 compared with 25 nonrefugee patients (8.19%) who tested positive for severe acute respiratory syndrome 2 (prevalence ratio, 2.16; 95% confidence interval, 1.04–4.51). In addition, 7 of the refugee women who tested positive for severe acute respiratory syndrome coronavirus 2 were from Central Africa. CONCLUSION The severe acute respiratory syndrome coronavirus 2 outbreak has disproportionately affected refugee populations. This study highlighted the utility of universal screening in mounting a rapid response to an evolving pandemic and how we can better serve refugee communities. Focused response may help achieve more equitable care related to severe acute respiratory syndrome 2 among vulnerable communities. The identification of such populations may help mitigate the spread of the disease and facilitate a timely, culturally, and linguistically enhanced public health response.
Collapse
Affiliation(s)
- Crista E Johnson-Agbakwu
- Refugee Women's Health Clinic, Valleywise Health, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Sood, Ali, Bhattarai, Chambliss, and Coonrod).,Arizona State University, Tempe, AZ (Dr Johnson-Agbakwu).,University of Arizona College of Medicine, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Ali, Bhattarai, and Coonrod).,Creighton University School of Medicine, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Ali, and Coonrod).,District Medical Group, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Sood, Ali, Chambliss, and Coonrod)
| | - Cortney M Eakin
- Refugee Women's Health Clinic, Valleywise Health, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Sood, Ali, Bhattarai, Chambliss, and Coonrod).,University of Arizona College of Medicine, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Ali, Bhattarai, and Coonrod).,Creighton University School of Medicine, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Ali, and Coonrod).,District Medical Group, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Sood, Ali, Chambliss, and Coonrod)
| | - Celeste V Bailey
- Refugee Women's Health Clinic, Valleywise Health, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Sood, Ali, Bhattarai, Chambliss, and Coonrod).,Creighton University School of Medicine, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Ali, and Coonrod).,St. Joseph's Hospital, Phoenix, AZ (Drs Bailey and Doehrman)
| | - Shelly Sood
- Refugee Women's Health Clinic, Valleywise Health, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Sood, Ali, Bhattarai, Chambliss, and Coonrod).,District Medical Group, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Sood, Ali, Chambliss, and Coonrod)
| | - Nyima Ali
- Refugee Women's Health Clinic, Valleywise Health, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Sood, Ali, Bhattarai, Chambliss, and Coonrod).,University of Arizona College of Medicine, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Ali, Bhattarai, and Coonrod).,Creighton University School of Medicine, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Ali, and Coonrod).,District Medical Group, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Sood, Ali, Chambliss, and Coonrod)
| | - Pooja Doehrman
- St. Joseph's Hospital, Phoenix, AZ (Drs Bailey and Doehrman)
| | - Bikash Bhattarai
- Refugee Women's Health Clinic, Valleywise Health, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Sood, Ali, Bhattarai, Chambliss, and Coonrod).,University of Arizona College of Medicine, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Ali, Bhattarai, and Coonrod)
| | - Linda Chambliss
- Refugee Women's Health Clinic, Valleywise Health, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Sood, Ali, Bhattarai, Chambliss, and Coonrod).,District Medical Group, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Sood, Ali, Chambliss, and Coonrod)
| | - Dean V Coonrod
- Refugee Women's Health Clinic, Valleywise Health, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Sood, Ali, Bhattarai, Chambliss, and Coonrod).,University of Arizona College of Medicine, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Ali, Bhattarai, and Coonrod).,Creighton University School of Medicine, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Bailey, Ali, and Coonrod).,District Medical Group, Phoenix, AZ (Drs Johnson-Agbakwu, Eakin, Sood, Ali, Chambliss, and Coonrod)
| |
Collapse
|
36
|
Yamada S, Fukushi S, Kinoshita H, Ohnishi M, Suzuki T, Fujimoto T, Saijo M, Maeda K. Assessment of SARS-CoV-2 infectivity of upper respiratory specimens from COVID-19 patients by virus isolation using VeroE6/TMPRSS2 cells. BMJ Open Respir Res 2021; 8:8/1/e000830. [PMID: 33627333 PMCID: PMC7907832 DOI: 10.1136/bmjresp-2020-000830] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/23/2022] Open
Abstract
Background An outbreak of novel coronavirus (SARS-CoV-2)-associated respiratory infectious diseases (COVID-19) emerged in 2019 and has spread rapidly in humans around the world. The demonstration of in vitro infectiousness of respiratory specimens is an informative surrogate for SARS-CoV-2 transmission from patients with COVID-19; accordingly, viral isolation assays in cell culture are an important aspect of laboratory diagnostics for COVID-19. Methods We developed a simple and rapid protocol for isolating SARS-CoV-2 from respiratory specimens using VeroE6/TMPRSS2 cells, a cell line that is highly susceptible to the virus. We also investigated a correlation between isolation of SARS-CoV-2 and viral load detected by real-time RT-PCR (rRT-PCR) using N2 primer/probe set that has been developed for testing of COVID-19 in Japan. Results The SARS-CoV-2 isolation protocol did not require blind passage of inoculated cells and yielded the results of viral isolation within 7 days after inoculation. Specimens with cycle threshold (Ct) values of <20.2, determined by rRT-PCR, were predicted to be isolation-positive. On the other hand, 6.9% of specimens with Ct values >35 were virus isolation-positive, indicating that low viral loads (high Ct values) in upper respiratory specimens do not always indicate no risk of containing transmissible virus. Conclusion In combination with rRT-PCR, the SARS-CoV-2 isolation protocol provides a means for assessing the potential risk of transmissible virus in upper respiratory specimens.
Collapse
Affiliation(s)
| | | | | | | | - Tadaki Suzuki
- Department of Pathology, NIID, Shinjuku-ku, Tokyo, Japan
| | - Tsuguto Fujimoto
- Center for Infectious Disease Risk Management, NIID, Shinjuku-ku, Tokyo, Japan
| | | | - Ken Maeda
- Department of Veterinary Science, NIID, Shinjuku-ku, Tokyo, Japan
| | | |
Collapse
|
37
|
Wadhwa A, Fisher KA, Silver R, Koh M, Arons MM, Miller DA, McIntyre AF, Vuong JT, Kim K, Takamiya M, Binder AM, Tate JE, Armstrong PA, Black SR, Mennella CC, Levin R, Gubser J, Jones B, Welbel SF, Moonan PK, Curran K, Ghinai I, Doshi R, Zawitz CJ. Identification of Presymptomatic and Asymptomatic Cases Using Cohort-Based Testing Approaches at a Large Correctional Facility-Chicago, Illinois, USA, May 2020. Clin Infect Dis 2021; 72:e128-e135. [PMID: 33270101 PMCID: PMC7799274 DOI: 10.1093/cid/ciaa1802] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/01/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) continues to cause significant morbidity and mortality worldwide. Correctional and detention facilities are at high risk of experiencing outbreaks. We aimed to evaluate cohort-based testing among detained persons exposed to laboratory-confirmed cases of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in order to identify presymptomatic and asymptomatic cases. METHODS During 1-19 May 2020, 2 testing strategies were implemented in 12 tiers or housing units of the Cook County Jail, Chicago, Illinois. Detained persons were approached to participate in serial testing (n = 137) and offered tests at 3 time points over 14 days (day 1, days 3-5, and days 13-14). The second group was offered a single test and interview at the end of a 14-day quarantine period (day 14 group) (n = 87). RESULTS 224 detained persons were approached for participation and, of these, 194 (87%) participated in ≥1 interview and 172 (77%) had ≥1 test. Of the 172 tested, 19 were positive for SARS-CoV-2. In the serial testing group, 17 (89%) new cases were detected, 16 (84%) on day 1, 1 (5%) on days 3-5, and none on days 13-14; in the day 14 group, 2 (11%) cases were identified. More than half (12/19; 63%) of the newly identified cases were presymptomatic or asymptomatic. CONCLUSIONS Our findings highlight the utility of cohort-based testing promptly after initiating quarantine within a housing tier. Cohort-based testing efforts identified new SARS-CoV-2 asymptomatic and presymptomatic infections that may have been missed by symptom screening alone.
Collapse
Affiliation(s)
- Ashutosh Wadhwa
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA.,Laboratory Leadership Service, CDC, Atlanta, Georgia, USA
| | - Kiva A Fisher
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA.,Epidemic Intelligence Service, CDC, Atlanta, Georgia, USA
| | - Rachel Silver
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA.,Public Health Institute-CDC Global Health Fellow, Atlanta, Georgia, USA
| | - Mitsuki Koh
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA.,Public Health Institute-CDC Global Health Fellow, Atlanta, Georgia, USA
| | - Melissa M Arons
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA.,Epidemic Intelligence Service, CDC, Atlanta, Georgia, USA
| | - David A Miller
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Anne F McIntyre
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Jeni T Vuong
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Kaylee Kim
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Mayuko Takamiya
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Alison M Binder
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Jacqueline E Tate
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Paige A Armstrong
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | | | - Connie C Mennella
- Cermak Health Services, Chicago, Illinois, USA.,Cook County Health, Chicago, Illinois, USA
| | - Rebecca Levin
- Cook County Sherriff's Office, Chicago, Illinois, USA
| | - Jane Gubser
- Cook County Sherriff's Office, Chicago, Illinois, USA
| | - Bridgette Jones
- Cermak Health Services, Chicago, Illinois, USA.,Cook County Health, Chicago, Illinois, USA
| | | | - Patrick K Moonan
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Kathryn Curran
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Isaac Ghinai
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA.,Epidemic Intelligence Service, CDC, Atlanta, Georgia, USA.,Chicago Department of Public Health, Chicago, Illinois, USA
| | - Reena Doshi
- COVID-19 Response Team, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Chad J Zawitz
- Cermak Health Services, Chicago, Illinois, USA.,Cook County Health, Chicago, Illinois, USA
| |
Collapse
|
38
|
Oley MH, Oley MC, Kepel BJ, Tjandra DE, Langi FLFG, Herwen, Aling DMR, Joicetine Wagiu AM, Faruk M. ICAM-1 levels in patients with covid-19 with diabetic foot ulcers: A prospective study in southeast asia. Ann Med Surg (Lond) 2021; 63:102171. [PMID: 33585030 PMCID: PMC7873613 DOI: 10.1016/j.amsu.2021.02.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Viral infection can compound the severity of pre-existing inflammation caused by underlying diseases. For those with a chronic, immune-related condition such as diabetic foot ulcers (DFUs), the coronavirus disease (COVID-19) serves to exacerbate the inflammatory burden. Serum levels of intracellular adhesion molecule-1 (ICAM-1), a primary mediator of cell adhesion express in the inflammatory process, are often used to indicate the gravity of all inflammatory conditions. Therefore, the purpose of this study was to investigate serum ICAM-1 levels before and after debridement in patients with DFUs who were also diagnosed as COVID-19 positive compared with those who were COVID-19 negative. METHODS 20 patients with DFUs were screened for COVID-19 and then divided into COVID-19 positive and negative groups according to the results. Before debridement, chest x-rays and blood analysis, including ICAM-1 serum levels, were performed in both groups. Only ICAM-1 serum levels were measured after debridement. RESULTS Of the 20 patients included in this study, 55% were male (n = 11) and 45% were female (n = 9). The mean age was 52.9 ± 1.9 years. ICAM-1 levels in patients with DFU in the COVID-19-positive group were significantly higher than those in the COVID-19-negative group (median 317.2 vs 149.2, respectively; p < 0.001). Serum levels of ICAM-1 reduced significantly in patients with DFU in the COVID-19-positive group were significantly higher than those in the COVID-19-negative group after debridement (median 312.5 vs 130.3; p < 0.001). CONCLUSION ICAM-1 serum levels represent an additional, initial screening marker for COVID-19.
Collapse
Affiliation(s)
- Mendy Hatibie Oley
- Plastic Reconstructive and Aesthetic Surgery Division, Department of Surgery, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
- Plastic Reconstructive and Aesthetic Surgery Division, Department of Surgery, R. D. Kandou Hospital, Manado, Indonesia
- Hyperbaric Centre Siloam Hospital, Manado, Indonesia
| | - Maximillian Christian Oley
- Hyperbaric Centre Siloam Hospital, Manado, Indonesia
- Neurosurgery Division, Department of Surgery, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
- Neurosurgery Division, Department of Surgery, R. D. Kandou Hospital, Manado, Indonesia
| | - Billy Johnson Kepel
- Department of Chemistry, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
| | - Djony E. Tjandra
- Hyperbaric Centre Siloam Hospital, Manado, Indonesia
- Vascular Surgery Division, Department of Surgery, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
- Vascular Surgery Division, Department of Surgery, R. D. Kandou Hospital, Manado, Indonesia
| | - Fima Lanra Fredrik G. Langi
- Department Epidemiology and Biostatistics, Public Health Faculty, Sam Ratulangi University, Manado, Indonesia
| | - Herwen
- Department of Surgery, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
- Department of Surgery, R. D. Kandou Hospital, Manado, Indonesia
| | | | - Angelica Maurene Joicetine Wagiu
- Department of Surgery, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
- Department of Surgery, R. D. Kandou Hospital, Manado, Indonesia
| | - Muhammad Faruk
- Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
39
|
Peng B, Zhou W, Pettit RW, Yu P, Matos PG, Greninger AL, McCashin J, Amos CI. Optimal test-assisted quarantine strategies for COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 33655268 DOI: 10.1101/2020.11.06.20222398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective To evaluate the effectiveness of SARS-CoV-2 testing on shortening the duration of quarantines for COVID-19 and to identify the most effective choices of testing schedules. Design We performed extensive simulations to evaluate the performance of quarantine strategies when one or more SARS-CoV-2 tests were administered during the quarantine. Simulations were based on statistical models for the transmissibility and viral loads of SARS-CoV-2 infections and the sensitivities of available testing methods. Sensitivity analyses were performed to evaluate the impact of perturbations in model assumptions on the outcomes of optimal strategies. Results We found that SARS-CoV-2 testing can effectively reduce the length of a quarantine without compromising safety. A single RT-PCR test performed before the end of quarantine can reduce quarantine duration to 10 days. Two tests can reduce the duration to 8 days, and three highly sensitive RT-PCR tests can justify a 6-day quarantine. More strategic testing schedules and longer quarantines are needed if tests are administered with less sensitive RT-PCR tests or antigen tests. Shorter quarantines can be utilized for applications that tolerate a residual post-quarantine transmission risk comparable to a 10-day quarantine. Conclusions Testing could substantially reduce the length of isolation, reducing the physical and mental stress caused by lengthy quarantines. With increasing capacity and lowered costs of SARS-CoV-2 tests, test-assisted quarantines could be safer and more cost-effective than 14-day quarantines and warrant more widespread use. RESEARCH IN CONTEXT What is already known on this topic?: Recommendations for quarantining individuals who could have been infected with COVID-19 are based on limited evidence.Despite recent theoretical and case studies of test-assisted quarantines, there has been no substantive investigation to quantify the safety and efficacy of, nor an exhaustive search for, optimal test-assisted quarantine strategies. What this study adds Our simulations indicate that the 14-day quarantine approach is overly conservative and can be safely shortened if testing is performed.Our recommendations include testing schedules that could be immediately adopted and implemented as government and industry policies. Role of the Funding Source A major technology company asked that we perform simulations to understand the optimal strategy for managing personnel quarantining before forming cohorts of individuals who would work closely together. The funding entity did not influence the scope or output of the study but requested that we include antigen testing as a component of the quarantining process. Patrick Yu and Peter Matos are employees of Corporate Medical Advisors, and International S.O.S employs Julie McCashin. Other funding sources are research grants and did not influence the investigation.
Collapse
|
40
|
Dubelbeiss E, Silverberg M, White C, Jaspan D, Goldberg J, Haines C. Repeat positive severe acute respiratory syndrome coronavirus 2 (coronavirus disease 2019) testing ≥90 days apart in pregnant women. Am J Obstet Gynecol MFM 2021; 3:100331. [PMID: 33618036 PMCID: PMC7893246 DOI: 10.1016/j.ajogmf.2021.100331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/07/2021] [Accepted: 02/10/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Emily Dubelbeiss
- Department of Obstetrics and Gynecology, Einstein Medical Center Philadelphia, Philadelphia, PA
| | - Michael Silverberg
- Department of Obstetrics and Gynecology, Einstein Medical Center Philadelphia, Philadelphia, PA
| | - Chase White
- Department of Obstetrics and Gynecology, Einstein Medical Center Philadelphia, Philadelphia, PA
| | - David Jaspan
- Department of Obstetrics and Gynecology, Einstein Medical Center Philadelphia, Philadelphia, PA
| | - Jay Goldberg
- Department of Obstetrics and Gynecology, Einstein Medical Center Philadelphia, Philadelphia, PA.
| | - Christopher Haines
- Department of Family and Community Medicine, Thomas Jefferson University Hospital, Philadelphia, PA
| |
Collapse
|
41
|
Analytical Sensitivity of the Abbott BinaxNOW COVID-19 Ag Card. J Clin Microbiol 2021; 59:JCM.02880-20. [PMID: 33310764 PMCID: PMC8106729 DOI: 10.1128/jcm.02880-20] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/10/2020] [Indexed: 12/03/2022] Open
Abstract
Multiple rapid antigen (Ag) tests for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have recently received emergency-use authorization (EUA) from the U.S. Food and Drug Administration (FDA). Multiple rapid antigen (Ag) tests for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have recently received emergency-use authorization (EUA) from the U.S. Food and Drug Administration (FDA). Although less sensitive than molecular detection methods, rapid antigen testing offers the potential for inexpensive, quick, decentralized testing. Robust analytical sensitivity data in comparison to reverse transcription-quantitative PCR (qRT-PCR) are currently lacking for many rapid antigen tests. Here, we evaluated the analytical sensitivity of the Abbott BinaxNOW COVID-19 Ag card using SARS-CoV-2-positive clinical specimens quantified by reverse transcription-droplet digital PCR (RT-ddPCR) and multiple FDA EUA qRT-PCR platforms using RNA standards. Initial and confirmatory limits of detection for the BinaxNOW COVID-19 Ag card were determined to be equivalent to 4.04 × 104 to 8.06 × 104 copies/swab. We further confirmed this limit of detection with 72 additional clinical samples positive for SARS-CoV-2 in either phosphate-buffered saline or viral transport medium. One hundred percent of samples with viral loads of >40,000 copies/swab were detected by rapid antigen testing. These data indicate that the BinaxNOW COVID-19 Ag card has an analytical sensitivity approximately equivalent to a generic qRT-PCR cycle threshold (CT) value of 29 to 30.
Collapse
|
42
|
Chaibun T, Puenpa J, Ngamdee T, Boonapatcharoen N, Athamanolap P, O'Mullane AP, Vongpunsawad S, Poovorawan Y, Lee SY, Lertanantawong B. Rapid electrochemical detection of coronavirus SARS-CoV-2. Nat Commun 2021; 12:802. [PMID: 33547323 PMCID: PMC7864991 DOI: 10.1038/s41467-021-21121-7] [Citation(s) in RCA: 267] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/11/2021] [Indexed: 12/20/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a highly contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Diagnosis of COVID-19 depends on quantitative reverse transcription PCR (qRT-PCR), which is time-consuming and requires expensive instrumentation. Here, we report an ultrasensitive electrochemical biosensor based on isothermal rolling circle amplification (RCA) for rapid detection of SARS-CoV-2. The assay involves the hybridization of the RCA amplicons with probes that were functionalized with redox active labels that are detectable by an electrochemical biosensor. The one-step sandwich hybridization assay could detect as low as 1 copy/μL of N and S genes, in less than 2 h. Sensor evaluation with 106 clinical samples, including 41 SARS-CoV-2 positive and 9 samples positive for other respiratory viruses, gave a 100% concordance result with qRT-PCR, with complete correlation between the biosensor current signals and quantitation cycle (Cq) values. In summary, this biosensor could be used as an on-site, real-time diagnostic test for COVID-19.
Collapse
Affiliation(s)
- Thanyarat Chaibun
- Biosensors Laboratory, Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand
| | - Jiratchaya Puenpa
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Tatchanun Ngamdee
- Department of Biotechnology, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Nimaradee Boonapatcharoen
- Pilot Plant Development and Training Institute (PDTI), King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Pornpat Athamanolap
- Biosensors Laboratory, Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand
| | - Anthony Peter O'Mullane
- School of Chemistry and Physics, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Sompong Vongpunsawad
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Su Yin Lee
- Faculty of Applied Sciences, AIMST University, Bedong, Kedah, Malaysia
- Centre of Excellence for Omics-Driven Computational Biodiscovery (COMBio), AIMST University, Bedong, Kedah, Malaysia
| | - Benchaporn Lertanantawong
- Biosensors Laboratory, Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand.
| |
Collapse
|
43
|
Yüce M, Filiztekin E, Özkaya KG. COVID-19 diagnosis -A review of current methods. Biosens Bioelectron 2021; 172:112752. [PMID: 33126180 PMCID: PMC7584564 DOI: 10.1016/j.bios.2020.112752] [Citation(s) in RCA: 398] [Impact Index Per Article: 99.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/01/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
A fast and accurate self-testing tool for COVID-19 diagnosis has become a prerequisite to comprehend the exact number of cases worldwide and to take medical and governmental actions accordingly. SARS-CoV-2 (formerly, 2019-nCoV) infection was first reported in Wuhan (China) in December 2019, and then it has rapidly spread around the world, causing ~14 million active cases with ~582,000 deaths as of July 2020. The diagnosis tools available so far have been based on a) viral gene detection, b) human antibody detection, and c) viral antigen detection, among which the viral gene detection by RT-PCR has been found as the most reliable technique. In this report, the current SARS-CoV-2 detection kits, exclusively the ones that were issued an "Emergency Use Authorization" from the U.S. Food and Drug Administration, were discussed. The key structural components of the virus were presented to provide the audience with an understanding of the scientific principles behind the testing tools. The methods that are still in the early research state were also reviewed in a subsection based on the reports available so far.
Collapse
Affiliation(s)
- Meral Yüce
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, 34956, Orhanli, Tuzla, Istanbul, Turkey.
| | - Elif Filiztekin
- Faculty of Engineering and Natural Sciences and, Sabanci University, 34956, Orhanli, Tuzla, Istanbul, Turkey
| | - Korin Gasia Özkaya
- Faculty of Engineering and Natural Sciences and, Sabanci University, 34956, Orhanli, Tuzla, Istanbul, Turkey
| |
Collapse
|
44
|
Bloom JA, Erlichman Z, Tian T, Doron S, Chen L, Mansur K, Tarnoff ME. The prevalence of asymptomatic carriers of COVID-19 as determined by routine preoperative testing. J Infect Prev 2021; 22:7-11. [PMID: 33841556 PMCID: PMC7841708 DOI: 10.1177/1757177420967096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/13/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The coronavirus disease SARS-CoV-2 (COVID-19) has swiftly spread throughout the globe, greatly influencing all aspects of life. As in previous pandemics, concerns for limited resources and a sustainable medical workforce have been on the forefront of infrastructure modifications. Consequently, surgical specialties have needed to consider each surgical case for necessity and safety during the COVID-19 outbreak. At our institution, availability of SARS-CoV-2 assay has allowed preoperative testing of asymptomatic surgical patients. AIM/OBJECTIVE To better define the prevalence of asymptomatic carriers in a surgical population and to better understand the impact of testing on our personal protective equipment (PPE) supply. METHODS We began routine, preoperative testing for all asymptomatic patients coming to our academic medical centre on 30 March 2020. Scheduled surgeries were deemed urgent by the surgeon with a review for appropriateness by a novel surgical committee. A retrospective patient chart review was performed. Emergency surgeries were excluded. Asymptomatic patients with positive test results had their surgeries rescheduled at the discretion of the surgeon and patient. Patients who tested negative underwent surgery with staff using standard PPE. RESULTS Eighty-four asymptomatic surgical patients were tested preoperatively with three (3.6%) testing positive for SARS-CoV-2. Preoperative testing saved 498 N95 respirators over this time period. DISCUSSION This is the first report of routine COVID-19 preoperative testing in an asymptomatic surgical population. Within this population, there is a 3.6% rate of asymptomatic SARS-CoV-2 carriers. Through this practice, personnel exposure can be minimised and access to PPE can be preserved.
Collapse
Affiliation(s)
| | | | - Tina Tian
- Tufts Medical Center, Boston, MA, USA
| | | | | | | | | |
Collapse
|
45
|
Prevalence of SARS-CoV-2 infection in patients presenting for intravitreal injection. SPEKTRUM DER AUGENHEILKUNDE 2020; 35:70-74. [PMID: 33343086 PMCID: PMC7731141 DOI: 10.1007/s00717-020-00473-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/18/2020] [Indexed: 02/04/2023]
Abstract
Aim Due to the coronavirus disease 2019 (COVID-19) pandemic, nosocomial transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is of great concern to clinicians of all specialties. Currently there are no published data available on the prevalence of the infection in ophthalmology patients presenting for intravitreal injection (IVI). The purpose of this retrospective study was to estimate the prevalence of SARS-CoV‑2 infection in patients presenting for IVI at our hospital. Methods Patients presenting for IVI in April 2020 at our hospital who had been screened for SARS-CoV‑2 infection using nasopharyngeal and oropharyngeal specimen for real-time reverse transcription polymerase chain reaction analysis were included in a retrospective study. To assess the representativity of this sample for IVI patients, characteristics were compared with patients presenting for IVI during March–April 2019. Results The study included 279 patients and 319 historic control patients. Of 277 valid test results, one SARS-CoV‑2 positive patient was found, resulting in a carrier rate of 0.36% with a 95% Clopper–Pearson confidence interval of 0.01–1.99%. No differences in sex (57.7% vs. 59.9% female, p = 0.650), age (77.63 ± 10.29 vs. 77.59 ± 10.94 years, p = 0.962), and region of residence were found between groups. Conclusion The study provides an estimate for the prevalence of SARS-CoV‑2 infection in asymptomatic patients presenting for IVI. While these data may be used as a baseline, further research is needed to assess the development of SARS-CoV‑2 prevalence in this patient group in order to support risk assessment and infection prevention strategies.
Collapse
|
46
|
Agarwal V, Venkatakrishnan AJ, Puranik A, Kirkup C, Lopez-Marquez A, Challener DW, Theel ES, O'Horo JC, Binnicker MJ, Kremers WK, Faubion WA, Badley AD, Williams AW, Gores GJ, Halamka JD, Morice WG, Soundararajan V. Long-term SARS-CoV-2 RNA shedding and its temporal association to IgG seropositivity. Cell Death Discov 2020; 6:138. [PMID: 33298894 PMCID: PMC7709096 DOI: 10.1038/s41420-020-00375-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/05/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022] Open
Abstract
Longitudinal characterization of SARS-CoV-2 PCR testing from COVID-19 patient's nasopharynx and its juxtaposition with blood-based IgG-seroconversion diagnostic assays is critical to understanding SARS-CoV-2 infection durations. Here, we retrospectively analyze 851 SARS-CoV-2-positive patients with at least two positive PCR tests and find that 99 of these patients remain SARS-CoV-2-positive after 4 weeks from their initial diagnosis date. For the 851-patient cohort, the mean lower bound of viral RNA shedding was 17.3 days (SD: 7.8), and the mean upper bound of viral RNA shedding from 668 patients transitioning to confirmed PCR-negative status was 22.7 days (SD: 11.8). Among 104 patients with an IgG test result, 90 patients were seropositive to date, with mean upper bound of time to seropositivity from initial diagnosis being 37.8 days (95% CI: 34.3-41.3). Our findings from juxtaposing IgG and PCR tests thus reveal that some SARS-CoV-2-positive patients are non-hospitalized and seropositive, yet actively shed viral RNA (14 of 90 patients). This study emphasizes the need for monitoring viral loads and neutralizing antibody titers in long-term non-hospitalized shedders as a means of characterizing the SARS-CoV-2 infection lifecycle.
Collapse
Affiliation(s)
- Vineet Agarwal
- nference, inc., One Main Street, Suite 400, East Arcade, Cambridge, MA, 02142, USA
| | - A J Venkatakrishnan
- nference, inc., One Main Street, Suite 400, East Arcade, Cambridge, MA, 02142, USA
| | - Arjun Puranik
- nference, inc., One Main Street, Suite 400, East Arcade, Cambridge, MA, 02142, USA
| | - Christian Kirkup
- nference, inc., One Main Street, Suite 400, East Arcade, Cambridge, MA, 02142, USA
| | | | | | | | | | | | | | | | | | | | | | | | - William G Morice
- Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Laboratories, Rochester, MN, USA
| | - Venky Soundararajan
- nference, inc., One Main Street, Suite 400, East Arcade, Cambridge, MA, 02142, USA.
| |
Collapse
|
47
|
Xu G, Emanuel AJ, Nadig S, Mehrotra S, Caddell BA, Curry SR, Nolte FS, Babic N. Evaluation of Orthogonal Testing Algorithm for Detection of SARS-CoV-2 IgG Antibodies. Clin Chem 2020; 66:1531-1537. [PMID: 32894753 PMCID: PMC7499512 DOI: 10.1093/clinchem/hvaa210] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/27/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibody testing is an important tool in assessment of pandemic progress, contact tracing, and identification of recovered coronavirus disease 2019 (COVID-19) patients. We evaluated an orthogonal testing algorithm (OTA) to improve test specificity in these use cases. METHODS A two-step OTA was applied where individuals who initially tested positive were tested with a second test. The first-line test, detecting IgG antibodies to the viral nucleocapsid protein, was validated in 130 samples and the second-line test, detecting IgG antibodies to the viral spike protein in 148 samples. The OTA was evaluated in 4333 clinical patient specimens. The seropositivity rates relative to the SARS-CoV-2 PCR positivity rates were evaluated from our entire patient population data (n = 5102). RESULTS The first-line test resulted in a clinical sensitivity of 96.4% (95% CI; 82.3% to 99.4%), and specificity of 99.0% (95% CI; 94.7% to 99.8%), whereas the second-line test had a sensitivity of 100% (95% CI; 87.1% to 100%) and specificity of 98.4% (95% CI; 94.2% to 99.5%). Using the OTA, 78/98 (80%) of initially positive SARS-CoV-2 IgG results were confirmed with a second-line test, while 11/42 (26%) of previously diagnosed COVID-19 patients had no detectable antibodies as long as 94 days post PCR diagnosis. CONCLUSION Our results show that an OTA can be used to identify patients who require further follow-up due to potential SARS CoV-2 IgG false positive results. In addition, serological testing may not be sufficiently sensitive to reliably detect prior COVID-19 infection.
Collapse
Affiliation(s)
- Gang Xu
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Anthony J Emanuel
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Satish Nadig
- Department of Surgery, Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | - Shikhar Mehrotra
- Department of Surgery, Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | - Brittany A Caddell
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Scott R Curry
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Frederick S Nolte
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Nikolina Babic
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
48
|
Knapp R, Tona V, Okada T, Sarpong R, Garg NK. Cyanoamidine Cyclization Approach to Remdesivir's Nucleobase. Org Lett 2020; 22:8430-8435. [PMID: 33085486 PMCID: PMC7653677 DOI: 10.1021/acs.orglett.0c03052] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Indexed: 11/30/2022]
Abstract
We report an alternative approach to the unnatural nucleobase fragment seen in remdesivir (Veklury). Remdesivir displays broad-spectrum antiviral activity and is currently being evaluated in Phase III clinical trials to treat patients with COVID-19. Our route relies on the formation of a cyanoamidine intermediate, which undergoes Lewis acid-mediated cyclization to yield the desired nucleobase. The approach is strategically distinct from prior routes and could further enable the synthesis of remdesivir and other small-molecule therapeutics.
Collapse
Affiliation(s)
- Rachel
R. Knapp
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, California 90095-1569, United States
| | - Veronica Tona
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, California 90095-1569, United States
| | - Taku Okada
- Department
of Chemistry, University of California, Berkeley, California 94720, United States
| | - Richmond Sarpong
- Department
of Chemistry, University of California, Berkeley, California 94720, United States
| | - Neil K. Garg
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, California 90095-1569, United States
| |
Collapse
|
49
|
Axell-House DB, Lavingia R, Rafferty M, Clark E, Amirian ES, Chiao EY. The estimation of diagnostic accuracy of tests for COVID-19: A scoping review. J Infect 2020; 81:681-697. [PMID: 32882315 PMCID: PMC7457918 DOI: 10.1016/j.jinf.2020.08.043] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 08/27/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVES To assess the methodologies used in the estimation of diagnostic accuracy of SARS-CoV-2 real-time reverse transcription polymerase chain reaction (rRT-PCR) and other nucleic acid amplification tests (NAATs) and to evaluate the quality and reliability of the studies employing those methods. METHODS We conducted a systematic search of English-language articles published December 31, 2019-June 19, 2020. Studies of any design that performed tests on ≥10 patients and reported or inferred correlative statistics were included. Studies were evaluated using elements of the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) guidelines. RESULTS We conducted a narrative and tabular synthesis of studies organized by their reference standard strategy or comparative agreement method, resulting in six categorizations. Critical study details were frequently unreported, including the mechanism for patient/sample selection and researcher blinding to results, which lead to concern for bias. CONCLUSIONS Current studies estimating test performance characteristics have imperfect study design and statistical methods for the estimation of test performance characteristics of SARS-CoV-2 tests. The included studies employ heterogeneous methods and overall have an increased risk of bias. Employing standardized guidelines for study designs and statistical methods will improve the process for developing and validating rRT-PCR and NAAT for the diagnosis of COVID-19.
Collapse
Affiliation(s)
- Dierdre B Axell-House
- Section of Infectious Diseases, Department of Internal Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Richa Lavingia
- Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; School of Social Sciences, Rice University, MS 272, 5620 Greenbriar Dr, Houston, TX 77005, USA; UTHealth School of Public Health, Houston, TX, USA
| | - Megan Rafferty
- School of Social Sciences, Rice University, MS 272, 5620 Greenbriar Dr, Houston, TX 77005, USA; UTHealth School of Public Health, Houston, TX, USA
| | - Eva Clark
- Section of Infectious Diseases, Department of Internal Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Houston HSR&D IQuESt, Michael E. DeBakey VA Medical Center, 2450 Holcombe Blvd, Houston, TX, 77021, USA
| | - E Susan Amirian
- School of Social Sciences, Rice University, MS 272, 5620 Greenbriar Dr, Houston, TX 77005, USA
| | - Elizabeth Y Chiao
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, 1155 Pressler St., Unit 1340, Houston, TX 77030, USA.
| |
Collapse
|
50
|
Gulholm T, Basile K, Kok J, Chen SCA, Rawlinson W. Laboratory diagnosis of severe acute respiratory syndrome coronavirus 2. Pathology 2020; 52:745-753. [PMID: 33131799 PMCID: PMC7543760 DOI: 10.1016/j.pathol.2020.09.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 12/20/2022]
Abstract
The first laboratory confirmed case of Coronavirus disease 2019 (COVID-19) in Australia was in Victoria on 25 January 2020 in a man returning from Wuhan city, Hubei province, the People's Republic of China. This was followed by three cases in New South Wales the following day. The Australian Government activated the Australian Health Sector Emergency Response Plan for Novel Coronavirus on 27 February 2020 in anticipation of a pandemic. Subsequently, the World Health Organization declared COVID-19 to be a Public Health Emergency of International Concern followed by a pandemic on 30 January 2020 and 11 March 2020, respectively. Laboratory testing for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for COVID-19, is key in identifying infected persons to guide timely public health actions of contact tracing and patient isolation to limit transmission of infection. This article aims to provide a comprehensive overview of current laboratory diagnostic methods for SARS-CoV-2, including nucleic acid testing, serology, rapid antigen detection and antibody tests, virus isolation and whole genome sequencing. The relative advantages and disadvantages of the different diagnostic tests are presented, as well as their value in different clinical, infection control and public health contexts. We also describe the challenges in the provision of SARS-CoV-2 diagnostics in Australia, a country with a relatively low COVID-19 incidence in the first pandemic wave but in which prevalence could rapidly change.
Collapse
Affiliation(s)
- T Gulholm
- Department of Infectious Diseases, Prince of Wales Hospital, Randwick, NSW, Australia.
| | - K Basile
- Centre for Infectious Diseases and Microbiology Laboratory Services, NSW Health Pathology - Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - J Kok
- Centre for Infectious Diseases and Microbiology Laboratory Services, NSW Health Pathology - Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia; Centre for Infectious Diseases and Microbiology - Public Health, Westmead Hospital, Westmead, NSW, Australia
| | - S C-A Chen
- Centre for Infectious Diseases and Microbiology Laboratory Services, NSW Health Pathology - Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia; Centre for Infectious Diseases and Microbiology - Public Health, Westmead Hospital, Westmead, NSW, Australia; Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney Westmead Hospital, Westmead, NSW, Australia
| | - W Rawlinson
- NSW Health Pathology, Serology and Virology Division, Prince of Wales Hospital, Randwick, NSW, Australia; Virology Research Laboratory, Prince of Wales Hospital, University of New South Wales, Randwick, NSW, Australia; School of Medical Sciences and School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Kensington, NSW, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Kensington, NSW, Australia
| |
Collapse
|