1
|
Zhapparova GA, Myrzakhmetova BS, Tlenchiyeva TM, Tussipova AA, Bissenbayeva KB, Toytanova AS, Kutumbetov LB. [Evaluation of the effectiveness of chemical inactivation and immunogenicity of the Omicron variant of the SARS-CoV-2 virus]. Vopr Virusol 2024; 69:459-469. [PMID: 39527768 DOI: 10.36233/0507-4088-253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION The rapid spread of coronavirus infection COVID-19 among the population of many countries around the world has contributed to the emergence of many genetic variants of SARS-CoV-2. Compared to previous coronavirus variants, the new Omicron variants have shown a noticeable degree of mutation. Virus inactivation is one of the most important steps in the development of inactivated vaccines. The chemical inactivation agents currently used are β-propiolactone and formaldehyde, but there is no uniform standard for designing and specifying the inactivation process. OBJECTIVE Evaluation and comparison of the effectiveness of chemical inactivation of two agents, formaldehyde and β-propiolactone against immunogenicity of the Omicron variant of the SARS-CoV-2 virus. MATERIALS AND METHODS Nasopharyngeal swabs were used to obtain the SARS-CoV-2 Omicron variant virus. Vero cell culture was used to isolate, reproduce, titrate the virus, and perform a neutralization reaction. The kinetics of studying the inactivation of the virus by chemical agents such as formaldehyde and β-propiolactone was carried out. RESULTS Studies have been conducted to comparatively evaluate the effectiveness of chemical agents used to inactivate the SARS-CoV-2 virus of the Omicron variant, planned for use in the production of an inactivated whole-virion vaccine. Formaldehyde and β-propiolactone were used as inactivation agents in concentrations of 0.05, 0.1, 0.5% of the total volume of the virus suspension. It has been established that complete inactivation of the virus by formaldehyde in the concentrations used at a temperature of 37 °C occurs within up to 2 hours, and when using beta-propiolactone, within up to 12 hours. CONCLUSION Inactivated virus samples have different antigenic activity depending on the concentration of the inactivation agents used. The most pronounced antigenic activity is manifested in samples of the pathogen that were treated with an inactivation agent at a mild concentration of 0.05%. Increasing the concentration of inactivation agent by 5 or more times leads to a significant decrease in the antigenicity of the SARS-CoV-2 virus. With the inactivation modes used, the loss of biological activity of the virus occurs faster and antigenicity is retained largely when treated with formaldehyde.
Collapse
|
2
|
Pinto G, Gelzo M, Cernera G, Esposito M, Illiano A, Serpico S, Pinchera B, Gentile I, Castaldo G, Amoresano A. Molecular fingerprint by omics-based approaches in saliva from patients affected by SARS-CoV-2 infection. JOURNAL OF MASS SPECTROMETRY : JMS 2024; 59:e5082. [PMID: 39228271 DOI: 10.1002/jms.5082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 06/10/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024]
Abstract
Clinical expression of coronavirus disease 2019 (COVID-19) infectionis widely variable including fatal cases and patients with mild symptoms and a rapid resolution. We studied saliva from 63 hospitalized COVID-19 patients and from 30 healthy controls by integrating large-scale proteomics, peptidomics and targeted metabolomics to assess the biochemical alterations following the infection and to obtain a set of putative biomarkers useful for noninvasive diagnosis. We used an untargeted approach by using liquid chromatography-tandem mass spectrometry (LC-MS/MS) for proteomics and peptidomics analysis and targeted LC-multiple reaction monitoring/MS for the analysis of amino acids. The levels of 77 proteins were significantly different in COVID-19 patients. Among these, seven proteins were found only in saliva from patients with COVID-19, four were up-regulated and three were down-regulated at least five-folds in saliva from COVID-19 patients in comparison to controls. The analysis of proteins revealed a complex balance between pro-inflammatory and anti-inflammatory proteins and a reduced amount of several proteins with immune activity that possibly favours the spreading of the virus. Such reduction could be related to the enhanced activity of endopeptidases induced by the infection that in turn caused an altered balance of free peptides. In fact, on a total of 28 peptides, 22 (80%) were differently expressed in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and control subjects. The multivariate analysis of such peptides permits to obtain a diagnostic algorithm that discriminate the two populations with a high diagnostic efficiency. Among amino acids, only threonine resulted significantly different between COVID-19 patients and controls, while alanine levels were significantly different between COVID-19 patients with different severity. In conclusion, the present study defined a set of molecules to be detected with a quick and easy method based on mass spectrometry tandem useful to reveal biochemical alterations involved in the pathogenesis of such a complex disease. Data are available via ProteomeXchange with identifier PXD045612.
Collapse
Affiliation(s)
- Gabriella Pinto
- Dipartimento di Scienze Chimiche, University of Naples Federico II, Naples, Italy
- Istituto Nazionale Biostrutture e Biosistemi-Consorzio Interuniversitario, Rome, Italy
| | - Monica Gelzo
- CEINGE-Biotecnologie avanzate Franco Salvatore, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples Federico II, Naples, Italy
| | - Gustavo Cernera
- CEINGE-Biotecnologie avanzate Franco Salvatore, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples Federico II, Naples, Italy
| | - Mariapia Esposito
- Dipartimento di Scienze Chimiche, University of Naples Federico II, Naples, Italy
| | - Anna Illiano
- Dipartimento di Scienze Chimiche, University of Naples Federico II, Naples, Italy
- Istituto Nazionale Biostrutture e Biosistemi-Consorzio Interuniversitario, Rome, Italy
| | - Stefania Serpico
- Dipartimento di Scienze Chimiche, University of Naples Federico II, Naples, Italy
| | - Biagio Pinchera
- Dipartimento di Medicina Clinica e Chirurgia, University of Naples Federico II, Naples, Italy
| | - Ivan Gentile
- Dipartimento di Medicina Clinica e Chirurgia, University of Naples Federico II, Naples, Italy
| | - Giuseppe Castaldo
- CEINGE-Biotecnologie avanzate Franco Salvatore, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples Federico II, Naples, Italy
| | - Angela Amoresano
- Dipartimento di Scienze Chimiche, University of Naples Federico II, Naples, Italy
- Istituto Nazionale Biostrutture e Biosistemi-Consorzio Interuniversitario, Rome, Italy
- CEINGE-Biotecnologie avanzate Franco Salvatore, Naples, Italy
| |
Collapse
|
3
|
Cutts T, Leung A, Banadyga L, Krishnan J. Inactivation Validation of Ebola, Marburg, and Lassa Viruses in AVL and Ethanol-Treated Viral Cultures. Viruses 2024; 16:1354. [PMID: 39339831 PMCID: PMC11436171 DOI: 10.3390/v16091354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
High-consequence pathogens such as the Ebola, Marburg, and Lassa viruses are handled in maximum-containment biosafety level 4 (BSL-4) laboratories. Genetic material is often isolated from such viruses and subsequently removed from BSL-4 laboratories for a multitude of downstream analyses using readily accessible technologies and equipment available at lower-biosafety level laboratories. However, it is essential to ensure that these materials are free of viable viruses before removal from BSL-4 laboratories to guarantee sample safety. This study details the in-house procedure used for validating the inactivation of Ebola, Marburg, and Lassa virus cultures after incubation with AVL lysis buffer (Qiagen) and ethanol. This study's findings show that no viable virus was detectable when high-titer cultures of Ebola, Marburg, and Lassa viruses were incubated with AVL lysis buffer for 10 min, followed by an equal volume of 95% ethanol for 3 min, using a method with a sensitivity of ≤0.8 log10 TCID50 as the limit of detection.
Collapse
Affiliation(s)
- Todd Cutts
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (T.C.); (A.L.); (L.B.)
| | - Anders Leung
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (T.C.); (A.L.); (L.B.)
| | - Logan Banadyga
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (T.C.); (A.L.); (L.B.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Jay Krishnan
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (T.C.); (A.L.); (L.B.)
| |
Collapse
|
4
|
Wei X, Wu J, Peng W, Chen X, Zhang L, Rong N, Yang H, Zhang G, Zhang G, Zhao B, Liu J. The Milk of Cows Immunized with Trivalent Inactivated Vaccines Provides Broad-Spectrum Passive Protection against Hand, Foot, and Mouth Disease in Neonatal Mice. Vaccines (Basel) 2024; 12:570. [PMID: 38932299 PMCID: PMC11209096 DOI: 10.3390/vaccines12060570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Hand, foot, and mouth disease (HFMD) is a contagious viral infection predominantly affecting infants and young children, caused by multiple enteroviruses, including Enterovirus 71 (EV71), Coxsackievirus A16 (CA16), Coxsackievirus A10 (CA10), and Coxsackievirus A6 (CA6). The high pathogenicity of HFMD has garnered significant attention. Currently, there is no specific treatment or broad-spectrum preventive measure available for HFMD, and existing monovalent vaccines have limited impact on the overall incidence or prevalence of the disease. Consequently, with the emergence of new viral strains driven by vaccine pressure, there is an urgent need to develop strategies for the rapid response and control of new outbreaks. In this study, we demonstrated the broad protective effect of maternal antibodies against three types of HFMD by immunizing mother mice with a trivalent inactivated vaccine targeting EV71, CA16, and CA10, using a neonatal mouse challenge model. Based on the feasibility of maternal antibodies as a form of passive immunization to prevent HFMD, we prepared a multivalent antiviral milk by immunizing dairy cows with the trivalent inactivated vaccine to target multiple HFMD viruses. In the neonatal mouse challenge model, this immunized milk exhibited extensive passive protection against oral infections caused by the three HFMD viruses. Compared to vaccines, this strategy may offer a rapid and broadly applicable approach to providing passive immunity for the prevention of HFMD, particularly in response to the swift emergence and spread of new variants.
Collapse
Affiliation(s)
- Xiaohui Wei
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China; (X.W.)
| | - Jing Wu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China; (X.W.)
| | - Wanjun Peng
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China; (X.W.)
| | - Xin Chen
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China; (X.W.)
| | - Lihong Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China; (X.W.)
| | - Na Rong
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China; (X.W.)
| | - Hekai Yang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China; (X.W.)
| | - Gengxin Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China; (X.W.)
| | - Gaoying Zhang
- Wuhan Servicebio Technology Co., Ltd., Wuhan 430079, China;
| | - Binbin Zhao
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China; (X.W.)
| | - Jiangning Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China; (X.W.)
| |
Collapse
|
5
|
Paull JS, Petros BA, Brock-Fisher TM, Jalbert SA, Selser VM, Messer KS, Dobbins ST, DeRuff KC, Deng D, Springer M, Sabeti PC. Optimisation and evaluation of viral genomic sequencing of SARS-CoV-2 rapid diagnostic tests: a laboratory and cohort-based study. THE LANCET. MICROBE 2024; 5:e468-e477. [PMID: 38621394 PMCID: PMC11322816 DOI: 10.1016/s2666-5247(23)00399-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 04/17/2024]
Abstract
BACKGROUND Sequencing of SARS-CoV-2 from rapid diagnostic tests (RDTs) can bolster viral genomic surveillance efforts; however, approaches to maximise and standardise pathogen genome recovery from RDTs remain underdeveloped. We aimed to systematically optimise the elution of genetic material from RDT components and to evaluate the efficacy of RDT sequencing for outbreak investigation. METHODS In this laboratory and cohort-based study we seeded RDTs with inactivated SARS-CoV-2 to optimise the elution of genomic material from RDT lateral flow strips. We measured the effect of changes in buffer type, time in buffer, and rotation on PCR cycle threshold (Ct) value. We recruited individuals older than 18 years residing in the greater Boston area, MA, USA, from July 18 to Nov 5, 2022, via email advertising to students and staff at Harvard University, MA, USA, and via broad social media advertising. All individuals recruited were within 5 days of a positive diagnostic test for SARS-CoV-2; no other relevant exclusion criteria were applied. Each individual completed two RDTs and one PCR swab. On Dec 29, 2022, we also collected RDTs from a convenience sample of individuals who were positive for SARS-CoV-2 and associated with an outbreak at a senior housing facility in MA, USA. We extracted all returned PCR swabs and RDT components (ie, swab, strip, or buffer); samples with a Ct of less than 40 were subject to amplicon sequencing. We compared the efficacy of elution and sequencing across RDT brands and components and used RDT-derived sequences to infer transmission links within the outbreak at the senior housing facility. We conducted metagenomic sequencing of negative RDTs from symptomatic individuals living in the senior housing facility. FINDINGS Neither elution duration of greater than 10 min nor rotation during elution impacted viral titres. Elution in Buffer AVL (Ct=31·4) and Tris-EDTA Buffer (Ct=30·8) were equivalent (p=0·34); AVL outperformed elution in lysis buffer and 50% lysis buffer (Ct=40·0, p=0·0029 for both) as well as Universal Viral Transport Medium (Ct=36·7, p=0·079). Performance of RDT strips was poorer than that of matched PCR swabs (mean Ct difference 10·2 [SD 4·3], p<0·0001); however, RDT swabs performed similarly to PCR swabs (mean Ct difference 4·1 [5·2], p=0·055). No RDT brand significantly outperformed another. Across sample types, viral load predicted the viral genome assembly length. We assembled greater than 80% complete genomes from 12 of 17 RDT-derived swabs, three of 18 strips, and four of 11 residual buffers. We generated outbreak-associated SARS-CoV-2 genomes using both amplicon and metagenomic sequencing and identified multiple introductions of the virus that resulted in downstream transmission. INTERPRETATION RDT-derived swabs are a reasonable alternative to PCR swabs for viral genomic surveillance and outbreak investigation. RDT-derived lateral flow strips yield accurate, but significantly fewer, viral reads than matched PCR swabs. Metagenomic sequencing of negative RDTs can identify viruses that might underlie patient symptoms. FUNDING The National Science Foundation, the Hertz Foundation, the National Institute of General Medical Sciences, Harvard Medical School, the Howard Hughes Medical Institute, the US Centers for Disease Control and Prevention, the Broad Institute and the National Institute of Allergy and Infectious Diseases.
Collapse
Affiliation(s)
- Jillian S Paull
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Brittany A Petros
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard Medical School and MIT, Cambridge, MA, USA; Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA, USA.
| | - Taylor M Brock-Fisher
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA; Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | | | | | | | | | | | - Davy Deng
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael Springer
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Pardis C Sabeti
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA; Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA; Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Harvard University, Boston, MA, USA.
| |
Collapse
|
6
|
Mak GC, Lau SS, Wong KK, Than EK, Ng AY, Hung DL. Optimizing heat inactivation for SARS-CoV-2 at 95 °C and its implications: A standardized approach. Heliyon 2024; 10:e28371. [PMID: 38655330 PMCID: PMC11035938 DOI: 10.1016/j.heliyon.2024.e28371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 11/20/2023] [Accepted: 03/18/2024] [Indexed: 04/26/2024] Open
Abstract
Background Standardized and validated heat inactivation procedure for Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are not available. For heat inactivation, various protocols were reported to prepare External Quality Assessment Programme (EQAP) samples without direct comparison between different durations. Objective To assess the heat inactivation procedures against SARS-CoV-2. The efficacy of the optimized condition was reflected by the results from laboratories testing the EQAP samples. Study design The SARS-CoV-2 strain was exposed to 95 °C in a water bath for three different time intervals, 5 min, 10 min and 15 min, respectively. The efficacy of inactivation was confirmed by the absence of cytopathic effects and decreasing viral load in 3 successive cell line passages. The viral stock inactivated by the optimal time interval was dispatched to EQAP participants and the result returned were analyzed. Results All of the three conditions were capable of inactivating the SARS-CoV-2 of viral load at around cycle threshold value of 10. When the 95 °C 10 min condition was chosen to prepare SARS-CoV-2 EQAP samples, they showed sufficient homogeneity and stability. High degree of consensus was observed among EQAP participants in all samples dispatched. Conclusions The conditions evaluated in the present study could be helpful for laboratories in preparing SARS-CoV-2 EQAP samples.
Collapse
Affiliation(s)
- Gannon C.K. Mak
- All from Microbiology Division, Public Health Laboratory Services Branch, Centre for Health Protection, Department of Health, Hong Kong Special Administrative Region
| | - Stephen S.Y. Lau
- All from Microbiology Division, Public Health Laboratory Services Branch, Centre for Health Protection, Department of Health, Hong Kong Special Administrative Region
| | - Kitty K.Y. Wong
- All from Microbiology Division, Public Health Laboratory Services Branch, Centre for Health Protection, Department of Health, Hong Kong Special Administrative Region
| | - Eunice K.Y. Than
- All from Microbiology Division, Public Health Laboratory Services Branch, Centre for Health Protection, Department of Health, Hong Kong Special Administrative Region
| | - Anita Y.Y. Ng
- All from Microbiology Division, Public Health Laboratory Services Branch, Centre for Health Protection, Department of Health, Hong Kong Special Administrative Region
| | - Derek L.L. Hung
- All from Microbiology Division, Public Health Laboratory Services Branch, Centre for Health Protection, Department of Health, Hong Kong Special Administrative Region
| |
Collapse
|
7
|
Chaki SP, Kahl-McDonagh MM, Neuman BW, Zuelke KA. Validating the inactivation of viral pathogens with a focus on SARS-CoV-2 to safely transfer samples from high-containment laboratories. Front Cell Infect Microbiol 2024; 14:1292467. [PMID: 38510962 PMCID: PMC10951993 DOI: 10.3389/fcimb.2024.1292467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/19/2024] [Indexed: 03/22/2024] Open
Abstract
Introduction Pathogen leak from a high-containment laboratory seriously threatens human safety, animal welfare, and environmental security. Transportation of pathogens from a higher (BSL4 or BSL3) to a lower (BSL2) containment laboratory for downstream experimentation requires complete pathogen inactivation. Validation of pathogen inactivation is necessary to ensure safety during transportation. This study established a validation strategy for virus inactivation. Methods SARS-CoV-2 wild type, delta, and omicron variants underwent heat treatment at 95°C for 10 minutes using either a hot water bath or a thermocycler. To validate the inactivation process, heat-treated viruses, and untreated control samples were incubated with A549-hACE2 and Vero E6-TMPRSS2-T2A-ACE2 cells. The cells were monitored for up to 72 hours for any cytopathic effects, visually and under a microscope, and for virus genome replication via RT-qPCR. The quality of post-treated samples was assessed for suitability in downstream molecular testing applications. Results Heat treatment at 95°C for 10 minutes effectively inactivated SARS-CoV-2 variants. The absence of cytopathic effects, coupled with the inability of virus genome replication, validated the efficacy of the inactivation process. Furthermore, the heat-treated samples proved to be qualified for COVID-19 antigen testing, RT-qPCR, and whole-genome sequencing. Discussion By ensuring the safety of sample transportation for downstream experimentation, this validation approach enhances biosecurity measures. Considerations for potential limitations, comparisons with existing inactivation methods, and broader implications of the findings are discussed.
Collapse
Affiliation(s)
- Sankar Prasad Chaki
- Global Health Research Complex, Division of Research, Texas A&M University, College Station, TX, United States
| | - Melissa M. Kahl-McDonagh
- Global Health Research Complex, Division of Research, Texas A&M University, College Station, TX, United States
| | - Benjamin W. Neuman
- Global Health Research Complex, Division of Research, Texas A&M University, College Station, TX, United States
- Department of Biological Sciences, Texas A&M University, College Station, TX, United States
- Department of Molecular Pathogenesis and Immunology, Texas A&M University, College Station, TX, United States
| | - Kurt A. Zuelke
- Global Health Research Complex, Division of Research, Texas A&M University, College Station, TX, United States
| |
Collapse
|
8
|
Voltan G, Antonelli G, Mondin A, Tizianel I, Sabbadin C, Barbot M, Basso D, Scaroni C, Ceccato F. Heat inactivation of SARS-CoV 2 enabled the measurement of salivary cortisol during COVID-19 pandemic. Endocrine 2024; 83:775-782. [PMID: 37991703 PMCID: PMC10901918 DOI: 10.1007/s12020-023-03597-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 11/03/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND AND AIM Salivary cortisol has become an essential tool in the management of cortisol-related disease. In 2020 the sudden outbreak of COVID-19 pandemic caused several concerns about the use of saliva, due to the risk of contamination, and a European consensus further discourage using salivary cortisol. To decrease infectious risk, we handled specimens by applying a heat treatment to inactivate viral particles, further evaluating the impact of the COVID-19 pandemic on the use of salivary cortisol in clinical practice. MATERIAL AND METHODS Saliva samples were exposed for 10 min at 70 °C, then cortisol was measured using LC-MS/MS. The number of salivary cortisol examinations from 2013 to 2022 was extracted from the local electronic database: those performed in 2019, 2020, and 2021 were analyzed and compared with the historical data. RESULTS During 2020 we observed a decrease of 408 (-20%) examinations (p = 0.05) compared to 2019; especially in salivary cortisol daily rhythm and salivary cortisol/cortisone ratio (respectively reduction of 47% and 88%, p = 0.003 and p = 0.001). Analyzing year 2021 compared with 2020 we reported an increase of 420 examinations (+20%, p = 0.01), with a complete recovery of salivary cortisol measurement (considering 2019: p = 0.71). Major differences were observed between morning salivary cortisol (-20%, p = 0.017), LNSC (-21%, p = 0.012) and salivary cortisol rhythm (-22%, p = 0.056). No Sars-Cov2 infections related to working exposure were reported among laboratory's employers. CONCLUSIONS We speculate that the adoption of an appropriate technique to inactivate viral particles in saliva specimens allowed the safety maintenance of salivary collections, also during the Sars-CoV-2 outbreak.
Collapse
Affiliation(s)
- Giacomo Voltan
- Department of Medicine DIMED, University of Padova, Padova, Italy
- Endocrine Disease Unit, University-Hospital of Padova, Padova, Italy
| | - Giorgia Antonelli
- Department of Medicine DIMED, University of Padova, Padova, Italy
- Laboratory Medicine Unit, University-Hospital of Padova, Padova, Italy
| | - Alessandro Mondin
- Department of Medicine DIMED, University of Padova, Padova, Italy
- Endocrine Disease Unit, University-Hospital of Padova, Padova, Italy
| | - Irene Tizianel
- Department of Medicine DIMED, University of Padova, Padova, Italy
- Endocrine Disease Unit, University-Hospital of Padova, Padova, Italy
| | - Chiara Sabbadin
- Department of Medicine DIMED, University of Padova, Padova, Italy
- Endocrine Disease Unit, University-Hospital of Padova, Padova, Italy
| | - Mattia Barbot
- Department of Medicine DIMED, University of Padova, Padova, Italy
- Endocrine Disease Unit, University-Hospital of Padova, Padova, Italy
| | - Daniela Basso
- Department of Medicine DIMED, University of Padova, Padova, Italy
- Laboratory Medicine Unit, University-Hospital of Padova, Padova, Italy
| | - Carla Scaroni
- Department of Medicine DIMED, University of Padova, Padova, Italy
- Endocrine Disease Unit, University-Hospital of Padova, Padova, Italy
| | - Filippo Ceccato
- Department of Medicine DIMED, University of Padova, Padova, Italy.
- Endocrine Disease Unit, University-Hospital of Padova, Padova, Italy.
| |
Collapse
|
9
|
Ding J, Xu X, Deng Y, Zheng X, Zhang T. Comparison of RT-ddPCR and RT-qPCR platforms for SARS-CoV-2 detection: Implications for future outbreaks of infectious diseases. ENVIRONMENT INTERNATIONAL 2024; 183:108438. [PMID: 38232505 DOI: 10.1016/j.envint.2024.108438] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
The increased frequency of human infectious disease outbreaks caused by RNA viruses worldwide in recent years calls for enhanced public health surveillance for better future preparedness. Wastewater-based epidemiology (WBE) is emerging as a valuable epidemiological tool for providing timely population-wide surveillance for disease prevention and response complementary to the current clinical surveillance system. Here, we compared the analytical performance and practical applications between predominant molecular detection methods of RT-qPCR and RT-ddPCR on SARS-CoV-2 detection in wastewater surveillance. When pure viral RNA was tested, RT-ddPCR exhibited superior quantification accuracy at higher concentration levels and achieved more sensitive detection with reduced variation at low concentration levels. Furthermore, RT-ddPCR consistently demonstrated more robust and accurate measurement either in the background of the wastewater matrix or with the presence of mismatches in the target regions of the consensus assay. Additionally, by detecting mock variant RNA samples, we found that RT-ddPCR outperformed RT-qPCR in virus genotyping by targeting specific loci with signature mutations in allele-specific (AS) assays, especially at low levels of allele frequencies and concentrations, which increased the possibility for sensitive low-prevalence variant detection in the population. Our study provides insights for detection method selection in the WBE applications for future infectious disease outbreaks.
Collapse
Affiliation(s)
- Jiahui Ding
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region
| | - Xiaoqing Xu
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region
| | - Yu Deng
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region
| | - Xiawan Zheng
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region
| | - Tong Zhang
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region.
| |
Collapse
|
10
|
Zhdanov DD, Ivin YY, Shishparenok AN, Kraevskiy SV, Kanashenko SL, Agafonova LE, Shumyantseva VV, Gnedenko OV, Pinyaeva AN, Kovpak AA, Ishmukhametov AA, Archakov AI. Perspectives for the creation of a new type of vaccine preparations based on pseudovirus particles using polio vaccine as an example. BIOMEDITSINSKAIA KHIMIIA 2023; 69:253-280. [PMID: 37937429 DOI: 10.18097/pbmc20236905253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Traditional antiviral vaccines are currently created by inactivating the virus chemically, most often using formaldehyde or β-propiolactone. These approaches are not optimal since they negatively affect the safety of the antigenic determinants of the inactivated particles and require additional purification stages. The most promising platforms for creating vaccines are based on pseudoviruses, i.e., viruses that have completely preserved the outer shell (capsid), while losing the ability to reproduce owing to the destruction of the genome. The irradiation of viruses with electron beam is the optimal way to create pseudoviral particles. In this review, with the example of the poliovirus, the main algorithms that can be applied to characterize pseudoviral particles functionally and structurally in the process of creating a vaccine preparation are presented. These algorithms are, namely, the analysis of the degree of genome destruction and coimmunogenicity. The structure of the poliovirus and methods of its inactivation are considered. Methods for assessing residual infectivity and immunogenicity are proposed for the functional characterization of pseudoviruses. Genome integrity analysis approaches, atomic force and electron microscopy, surface plasmon resonance, and bioelectrochemical methods are crucial to structural characterization of the pseudovirus particles.
Collapse
Affiliation(s)
- D D Zhdanov
- Institute of Biomedical Chemistry, Moscow, Russia
| | - Yu Yu Ivin
- Institute of Biomedical Chemistry, Moscow, Russia; Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow, Russia
| | | | | | | | | | - V V Shumyantseva
- Institute of Biomedical Chemistry, Moscow, Russia; Pirogov Russian National Research Medical University, Moscow, Russia
| | - O V Gnedenko
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A N Pinyaeva
- Institute of Biomedical Chemistry, Moscow, Russia; Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow, Russia
| | - A A Kovpak
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A A Ishmukhametov
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow, Russia
| | - A I Archakov
- Institute of Biomedical Chemistry, Moscow, Russia; Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
11
|
Das A, Ahmed Z, Xu L, Jia W. Assessment and verification of chemical inactivation of peste des petits ruminants virus by virus isolation following virus capture using Nanotrap magnetic virus particles. Microbiol Spectr 2023; 11:e0068923. [PMID: 37655907 PMCID: PMC10580900 DOI: 10.1128/spectrum.00689-23] [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: 02/16/2023] [Accepted: 06/28/2023] [Indexed: 09/02/2023] Open
Abstract
IMPORTANCE Research including diagnosis on highly contagious viruses at the molecular level such as PCR and next-generation sequencing requires complete inactivation of the virus to ensure biosafety and biosecurity so that any accidental release of the virus does not compromise the safety of the susceptible population and the environment. In this work, peste des petits ruminants virus (PPRV) was inactivated with chemical agents, and the virus inactivation was confirmed by virus isolation (VI) using Vero cells. Since the chemical agents are cytotoxic, inactivated virus (PPRV) was diluted 1:100 to neutralize cytotoxicity, and the residual viruses (if any) were captured using Nanotrap magnetic virus particles (NMVPs). The NMVPs and the captured viruses were subjected to VI. No CPE was observed, indicating complete inactivation, and the results were further supported by real-time RT-PCR. This new protocol to verify virus inactivation can be applicable to other viruses.
Collapse
Affiliation(s)
- Amaresh Das
- US Department of Agriculture, Animal and Plant Health Inspection Service, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory, Reagents and Vaccine Services Section, Plum Island Animal Disease Center, Orient Point, New York, USA
| | - Zaheer Ahmed
- US Department of Agriculture, Animal and Plant Health Inspection Service, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory, Reagents and Vaccine Services Section, Plum Island Animal Disease Center, Orient Point, New York, USA
| | - Lizhe Xu
- US Department of Agriculture, Animal and Plant Health Inspection Service, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory, Reagents and Vaccine Services Section, Plum Island Animal Disease Center, Orient Point, New York, USA
| | - Wei Jia
- US Department of Agriculture, Animal and Plant Health Inspection Service, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory, Reagents and Vaccine Services Section, Plum Island Animal Disease Center, Orient Point, New York, USA
| |
Collapse
|
12
|
Boukli N, Flamand C, Chea KL, Heng L, Keo S, Sour K, In S, Chhim P, Chhor B, Kruy L, Feenstra JDM, Gandhi M, Okafor O, Ulekleiv C, Auerswald H, Horm VS, Karlsson EA. One assay to test them all: Multiplex assays for expansion of respiratory virus surveillance. Front Med (Lausanne) 2023; 10:1161268. [PMID: 37168265 PMCID: PMC10165998 DOI: 10.3389/fmed.2023.1161268] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/24/2023] [Indexed: 05/13/2023] Open
Abstract
Molecular multiplex assays (MPAs) for simultaneous detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza and respiratory syncytial virus (RSV) in a single RT-PCR reaction reduce time and increase efficiency to identify multiple pathogens with overlapping clinical presentation but different treatments or public health implications. Clinical performance of XpertXpress® SARS-CoV-2/Flu/RSV (Cepheid, GX), TaqPath™ COVID-19, FluA/B, RSV Combo kit (Thermo Fisher Scientific, TP), and PowerChek™ SARS-CoV-2/Influenza A&B/RSV Multiplex RT-PCR kit II (KogeneBiotech, PC) was compared to individual Standards of Care (SoC). Thirteen isolates of SARS-CoV-2, human seasonal influenza, and avian influenza served to assess limit of detection (LoD). Then, positive and negative residual nasopharyngeal specimens, collected under public health surveillance and pandemic response served for evaluation. Subsequently, comparison of effectiveness was assessed. The three MPAs confidently detect all lineages of SARS-CoV-2 and influenza viruses. MPA-LoDs vary from 1 to 2 Log10 differences from SoC depending on assay and strain. Clinical evaluation resulted in overall agreement between 97 and 100%, demonstrating a high accuracy to detect all targets. Existing differences in costs, testing burden and implementation constraints influence the choice in primary or community settings. TP, PC and GX, reliably detect SARS-CoV-2, influenza and RSV simultaneously, with reduced time-to-results and simplified workflows. MPAs have the potential to enhance diagnostics, surveillance system, and epidemic response to drive policy on prevention and control of viral respiratory infections.
Collapse
Affiliation(s)
- Narjis Boukli
- Virology Unit, National Influenza Center, WHO H5 Regional Reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Claude Flamand
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, CNRS, Paris, France
| | - Kim Lay Chea
- Virology Unit, National Influenza Center, WHO H5 Regional Reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Leangyi Heng
- Virology Unit, National Influenza Center, WHO H5 Regional Reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Seangmai Keo
- Virology Unit, National Influenza Center, WHO H5 Regional Reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Kimhoung Sour
- Virology Unit, National Influenza Center, WHO H5 Regional Reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sophea In
- Virology Unit, National Influenza Center, WHO H5 Regional Reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Panha Chhim
- Virology Unit, National Influenza Center, WHO H5 Regional Reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Bunthea Chhor
- Virology Unit, National Influenza Center, WHO H5 Regional Reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Lomor Kruy
- Virology Unit, National Influenza Center, WHO H5 Regional Reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | | | - Manoj Gandhi
- Thermo Fisher Scientific, South San Francisco CA, United States
| | - Obiageli Okafor
- Thermo Fisher Scientific, South San Francisco CA, United States
| | | | - Heidi Auerswald
- Virology Unit, National Influenza Center, WHO H5 Regional Reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Viseth Srey Horm
- Virology Unit, National Influenza Center, WHO H5 Regional Reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Erik A. Karlsson
- Virology Unit, National Influenza Center, WHO H5 Regional Reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| |
Collapse
|
13
|
Olejnik J, Leon J, Michelson D, Chowdhary K, Galvan-Pena S, Benoist C, Mühlberger E, Hume AJ. Establishment of an Inactivation Method for Ebola Virus and SARS-CoV-2 Suitable for Downstream Sequencing of Low Cell Numbers. Pathogens 2023; 12:342. [PMID: 36839614 PMCID: PMC9958562 DOI: 10.3390/pathogens12020342] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Technologies that facilitate the bulk sequencing of small numbers of cells as well as single-cell RNA sequencing (scRNA-seq) have aided greatly in the study of viruses as these analyses can be used to differentiate responses from infected versus bystander cells in complex systems, including in organoid or animal studies. While protocols for these analyses are typically developed with biosafety level 2 (BSL-2) considerations in mind, such analyses are equally useful for the study of viruses that require higher biosafety containment levels. Many of these workstreams, however, are not directly compatible with the more stringent biosafety regulations of BSL-3 and BSL-4 laboratories ensuring virus inactivation and must therefore be modified. Here we show that TCL buffer (Qiagen), which was developed for bulk sequencing of small numbers of cells and also facilitates scRNA-seq, inactivates both Ebola virus (EBOV) and SARS-CoV-2, BSL-4 and BSL-3 viruses, respectively. We show that additional heat treatment, necessary for the more stringent biosafety concerns for BSL-4-derived samples, was additionally sufficient to inactivate EBOV-containing samples. Critically, this heat treatment had minimal effects on extracted RNA quality and downstream sequencing results.
Collapse
Affiliation(s)
- Judith Olejnik
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02218, USA
| | - Juliette Leon
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- INSERM UMR 1163, Institut Imagine, University of Paris, 75015 Paris, France
| | - Daniel Michelson
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Kaitavjeet Chowdhary
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Silvia Galvan-Pena
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Christophe Benoist
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Elke Mühlberger
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02218, USA
| | - Adam J. Hume
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02218, USA
- Center for Emerging Infectious Diseases Policy & Research, Boston University, Boston, MA 02118, USA
| |
Collapse
|
14
|
BOUKLI N, FLAMAND C, CHEA KL, HENG L, KEO S, SOUR K, IN S, CHHIM P, CHHOR B, KRUY L, FEENSTRA JDM, GANDHI M, OKAFOR O, ULEKLIEV C, AUERSWALD H, HORM VS, KARLSSON EA. ONE ASSAY TO TEST THEM ALL: COMPARING MULTIPLEX ASSAYS FOR EXPANSION OF RESPIRATORY VIRUS SURVEILLANCE. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.19.23284806. [PMID: 36711477 PMCID: PMC9882628 DOI: 10.1101/2023.01.19.23284806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Background Molecular multiplex assays (MPAs) for simultaneous detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza and respiratory syncytial virus (RSV) in a single RT-PCR reaction reduce time and increase efficiency to identify multiple pathogens with overlapping clinical presentation but different treatments or public health implications. Methods Clinical performance of XpertXpress ® SARS-CoV-2/Flu/RSV (Cepheid, GX), TaqPath™ COVID-19, FluA/B, RSV Combo kit (Thermo Fisher Scientific, TP), and PowerChek™ SARS-CoV-2/Influenza A&B/RSV Multiplex RT-PCR kit II (KogeneBiotech, PC) was compared to individual Standards of Care (SoC). Thirteen isolates of SARS-CoV-2, human seasonal influenza, and avian influenza served to assess limit of detection (LoD). Then, positive and negative residual nasopharyngeal specimens, collected under public health surveillance and pandemic response served for evaluation. Subsequently, comparison of effectiveness was assessed. Results The three MPAs confidently detect all lineages of SARS-CoV-2 and influenza viruses. MPA-LoDs vary from 1-2 Log10 differences from SoC depending on assay and strain. Clinical evaluation resulted in overall agreement between 97% and 100%, demonstrating a high accuracy to detect all targets. Existing differences in costs, testing burden and implementation constraints influence the choice in primary or community settings. Conclusion TP, PC and GX, reliably detect SARS-CoV-2, influenza and RSV simultaneously, with reduced time-to-results and simplified workflows. MPAs have the potential to enhancediagnostics, surveillance system, and epidemic response to drive policy on prevention and control of viral respiratory infections. IMPORTANCE Viral respiratory infections represent a major burden globally, weighed down by the COVID-19 pandemic, and threatened by spillover of novel zoonotic influenza viruses. Since respiratory infections share clinical presentations, identification of the causing agent for patient care and public health measures requires laboratory testing for several pathogens, including potential zoonotic spillovers. Simultaneous detection of SARS-CoV-2, influenza, and RSV in a single RT-PCR accelerates time from sampling to diagnosis, preserve consumables, and streamline human resources to respond to other endemic or emerging pathogens. Multiplex assays have the potential to sustain and even expand surveillance systems, can utilize capacity/capability developed during the COVID-19 pandemic worldwide, thereby strengthening epidemic/pandemic preparedness, prevention, and response.
Collapse
Affiliation(s)
- Narjis BOUKLI
- Virology Unit, National Influenza Center, WHO H5 Regional reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Claude FLAMAND
- Epidemiology Unit, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Kim Lay CHEA
- Virology Unit, National Influenza Center, WHO H5 Regional reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Leangyi HENG
- Virology Unit, National Influenza Center, WHO H5 Regional reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Seangmai KEO
- Virology Unit, National Influenza Center, WHO H5 Regional reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Kimhoung SOUR
- Virology Unit, National Influenza Center, WHO H5 Regional reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sophea IN
- Virology Unit, National Influenza Center, WHO H5 Regional reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Panha CHHIM
- Virology Unit, National Influenza Center, WHO H5 Regional reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Bunthea CHHOR
- Virology Unit, National Influenza Center, WHO H5 Regional reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Lomor KRUY
- Virology Unit, National Influenza Center, WHO H5 Regional reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | | | - Manoj GANDHI
- Thermo Fisher Scientific, South San Francisco CA, United States
| | - Obiageli OKAFOR
- Thermo Fisher Scientific, South San Francisco CA, United States
| | | | - Heidi AUERSWALD
- Virology Unit, National Influenza Center, WHO H5 Regional reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Viseth Srey HORM
- Virology Unit, National Influenza Center, WHO H5 Regional reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Erik A KARLSSON
- Virology Unit, National Influenza Center, WHO H5 Regional reference Laboratory, World Health Organization COVID-19 Global Referral Laboratory, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| |
Collapse
|
15
|
Cimolai N. Disinfection and decontamination in the context of SARS-CoV-2-specific data. J Med Virol 2022; 94:4654-4668. [PMID: 35758523 PMCID: PMC9350315 DOI: 10.1002/jmv.27959] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/13/2022] [Accepted: 06/24/2022] [Indexed: 11/08/2022]
Abstract
Given the high transmissibility of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as witnessed early in the coronavirus disease 2019 (COVID-19) pandemic, concerns arose with the existing methods for virus disinfection and decontamination. The need for SARS-CoV-2-specific data stimulated considerable research in this regard. Overall, SARS-CoV-2 is practically and equally susceptible to approaches for disinfection and decontamination that have been previously found for other human or animal coronaviruses. The latter have included techniques utilizing temperature modulation, pH extremes, irradiation, and chemical treatments. These physicochemical methods are a necessary adjunct to other prevention strategies, given the environmental and patient surface ubiquity of the virus. Classic studies of disinfection have also allowed for extrapolation to the eradication of the virus on human mucosal surfaces by some chemical means. Despite considerable laboratory study, practical field assessments are generally lacking and need to be encouraged to confirm the correlation of interventions with viral eradication and infection prevention. Transparency in the constitution and use of any method or chemical is also essential to furthering practical applications.
Collapse
Affiliation(s)
- Nevio Cimolai
- Department of Pathology and Laboratory Medicine, Faculty of MedicineThe University of British ColumbiaVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineChildren's and Women's Health Centre of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
16
|
Stokes W, Berenger BM, Venner AA, Deslandes V, Shaw JLV. Point of care molecular and antigen detection tests for COVID-19: current status and future prospects. Expert Rev Mol Diagn 2022; 22:797-809. [PMID: 36093682 DOI: 10.1080/14737159.2022.2122712] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Detection of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) has been critical to support and management of the COVID-19 pandemic. Point of care testing (POCT) for SARS-CoV-2 has been a widely used tool for detection of SARS-CoV-2. AREAS COVERED POCT nucleic acid amplification tests (NAATs) and rapid antigen tests (RATs) have been the most readily used POCT for SARS-CoV-2. Here, current knowledge on the utility of POCT NAATs and RATs for SARS-CoV-2 are reviewed and discussed alongside aspects of quality assurance factors that must be considered for successful and safe implementation of POCT. EXPERT OPINION Use cases for implementation of POCT must be evidence based, regardless of the test used. A quality assurance framework must be in place to ensure accuracy and safety of POCT.
Collapse
Affiliation(s)
- William Stokes
- Alberta Precision Laboratories, Alberta, Canada.,Department of Pathology and Laboratory Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Byron M Berenger
- Alberta Precision Laboratories, Alberta, Canada.,Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Allison A Venner
- Alberta Precision Laboratories, Alberta, Canada.,Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Vincent Deslandes
- Eastern Ontario Regional Laboratories Association, Ottawa, Ontario, Canada.,Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada.,Department of Pathology and Laboratory Medicine, The University of Ottawa, Ottawa, Ontario, Canada
| | - Julie L V Shaw
- Eastern Ontario Regional Laboratories Association, Ottawa, Ontario, Canada.,Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada.,Department of Pathology and Laboratory Medicine, The University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
17
|
Yu S, Wei Y, Liang H, Ji W, Chang Z, Xie S, Wang Y, Li W, Liu Y, Wu H, Li J, Wang H, Yang X. Comparison of Physical and Biochemical Characterizations of SARS-CoV-2 Inactivated by Different Treatments. Viruses 2022; 14:v14091938. [PMID: 36146745 PMCID: PMC9503440 DOI: 10.3390/v14091938] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 08/27/2022] [Accepted: 08/28/2022] [Indexed: 12/02/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused huge social and economic distress. Given its rapid spread and the lack of specific treatment options, SARS-CoV-2 needs to be inactivated according to strict biosafety measures during laboratory diagnostics and vaccine development. The inactivation method for SARS-CoV-2 affects research related to the natural virus and its immune activity as an antigen in vaccines. In this study, we used size exclusion chromatography, western blotting, ELISA, an electron microscope, dynamic light scattering, circular dichroism, and surface plasmon resonance to evaluate the effects of four different chemical inactivation methods on the physical and biochemical characterization of SARS-CoV-2. Formaldehyde and β-propiolactone (BPL) treatment can completely inactivate the virus and have no significant effects on the morphology of the virus. None of the four tested inactivation methods affected the secondary structure of the virus, including the α-helix, antiparallel β-sheet, parallel β-sheet, β-turn, and random coil. However, formaldehyde and long-term BPL treatment (48 h) resulted in decreased viral S protein content and increased viral particle aggregation, respectively. The BPL treatment for 24 h can completely inactivate SARS-CoV-2 with the maximum retention of the morphology, physical properties, and the biochemical properties of the potential antigens of the virus. In summary, we have established a characterization system for the comprehensive evaluation of virus inactivation technology, which has important guiding significance for the development of vaccines against SARS-CoV-2 variants and research on natural SARS-CoV-2.
Collapse
Affiliation(s)
- Shouzhi Yu
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China
| | - Yangyang Wei
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China
| | - Hongyang Liang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China
| | - Wenheng Ji
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China
| | - Zhen Chang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China
| | - Siman Xie
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China
| | - Yichuan Wang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China
| | - Wanli Li
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China
| | - Yingwei Liu
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China
| | - Hao Wu
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China
| | - Jie Li
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China
| | - Hui Wang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China
- Correspondence: (H.W.); (X.Y.)
| | - Xiaoming Yang
- China National Biotec Group Company Limited, Beijing 100024, China
- Correspondence: (H.W.); (X.Y.)
| |
Collapse
|
18
|
Auerswald H, Low DHW, Siegers JY, Ou T, Kol S, In S, Linster M, Su YCF, Mendenhall IH, Duong V, Smith GJD, Karlsson EA. A Look inside the Replication Dynamics of SARS-CoV-2 in Blyth's Horseshoe Bat ( Rhinolophus lepidus) Kidney Cells. Microbiol Spectr 2022; 10:e0044922. [PMID: 35638834 PMCID: PMC9241725 DOI: 10.1128/spectrum.00449-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/11/2022] [Indexed: 01/09/2023] Open
Abstract
Bats are considered the natural reservoir of numerous emerging viruses such as severe acute respiratory syndrome coronaviruses (SARS-CoVs). There is a need for immortalized bat cell lines to culture and investigate the pathogenicity, replication kinetics, and evolution of emerging coronaviruses. We illustrate the susceptibility and permissiveness of a spontaneously immortalized kidney cell line (Rhileki) from Blyth's horseshoe bat (R. lepidus) to SARS-CoV-2 virus, including clinical isolates, suggesting a possible virus-host relationship. We were able to observe limited SARS-CoV-2 replication in Rhileki cells compared with simian VeroE6 cells. Slower viral replication in Rhileki cells was indicated by higher ct values (RT-PCR) at later time points of the viral culture and smaller foci (foci forming assay) compared with those of VeroE6 cells. With this study we demonstrate that SARS-CoV-2 replication is not restricted to R. sinicus and could include more Rhinolophus species. The establishment of a continuous Rhinolophus lepidus kidney cell line allows further characterization of SARS-CoV-2 replication in Rhinolophus bat cells, as well as isolation attempts of other bat-borne viruses. IMPORTANCE The current COVID-19 pandemic demonstrates the significance of bats as reservoirs for severe viral diseases. However, as bats are difficult to establish as animal models, bat cell lines can be an important proxy for the investigation of bat-virus interactions and the isolation of bat-borne viruses. This study demonstrates the susceptibility and permissiveness of a continuous kidney bat cell line to SARS-CoV-2. This does not implicate the bat species Rhinolophus lepidus, where these cells originate from, as a potential reservoir, but emphasizes the usefulness of this cell line for further characterization of SARS-CoV-2. This can lead to a better understanding of emerging viruses that could cause significant disease in humans and domestic animals.
Collapse
Affiliation(s)
- Heidi Auerswald
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Dolyce H. W. Low
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Jurre Y. Siegers
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Teyputita Ou
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Sonita Kol
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Saraden In
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Martin Linster
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Yvonne C. F. Su
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Ian H. Mendenhall
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- SingHealth Duke-NUS Global Health Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Gavin J. D. Smith
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- SingHealth Duke-NUS Global Health Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore
- Duke Global Health Institute, Duke University, Durham, North Carolina, USA
| | - Erik A. Karlsson
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| |
Collapse
|
19
|
Zheng X, Deng Y, Xu X, Li S, Zhang Y, Ding J, On HY, Lai JCC, In Yau C, Chin AWH, Poon LLM, Tun HM, Zhang T. Comparison of virus concentration methods and RNA extraction methods for SARS-CoV-2 wastewater surveillance. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 824:153687. [PMID: 35134418 PMCID: PMC8816846 DOI: 10.1016/j.scitotenv.2022.153687] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/16/2022] [Accepted: 02/01/2022] [Indexed: 05/02/2023]
Abstract
Wastewater surveillance is a promising tool for population-level monitoring of the spread of infectious diseases, such as the coronavirus disease 2019 (COVID-19). Different from clinical specimens, viruses in community-scale wastewater samples need to be concentrated before detection because viral RNA is highly diluted. The present study evaluated eleven different virus concentration methods for the detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in wastewater. First, eight concentration methods of different principles were compared using spiked wastewater at a starting volume of 30 mL. Ultracentrifugation was the most effective method with a viral recovery efficiency of 25 ± 6%. The second-best option, AlCl3 precipitation method, yielded a lower recovery efficiency, only approximately half that of the ultracentrifugation method. Second, the potential of increasing method sensitivity was explored using three concentration methods starting with a larger volume of 1000 mL. Although ultracentrifugation using a large volume outperformed the other two large-volume methods, it only yielded a comparable method sensitivity as the ultracentrifugation using a small volume (30 mL). Thus, ultracentrifugation using less volume of wastewater is more preferable considering the sample processing throughput. Third, a comparison of two viral RNA extraction methods showed that the lysis-buffer-based extraction method resulted in higher viral recovery efficiencies, with cycle threshold (Ct) values 0.9-4.2 lower than those obtained for the acid-guanidinium-phenol-based method using spiked samples. These results were further confirmed by using positive wastewater samples concentrated by ultracentrifugation and extracted separately by the two viral RNA extraction methods. In summary, concentration using ultracentrifugation followed by the lysis buffer-based extraction method enables sensitive and robust detection of SARS-CoV-2 for wastewater surveillance.
Collapse
Affiliation(s)
- Xiawan Zheng
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Yu Deng
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Xiaoqing Xu
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Shuxian Li
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Yulin Zhang
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Jiahui Ding
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Hei Yin On
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong, China; HKU-Pasteur Research Pole, Pokfulam Road, Hong Kong, China
| | - Jimmy C C Lai
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong, China; HKU-Pasteur Research Pole, Pokfulam Road, Hong Kong, China
| | - Chung In Yau
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Alex W H Chin
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Leo L M Poon
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong, China; HKU-Pasteur Research Pole, Pokfulam Road, Hong Kong, China
| | - Hein M Tun
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong, China; HKU-Pasteur Research Pole, Pokfulam Road, Hong Kong, China
| | - Tong Zhang
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China.
| |
Collapse
|
20
|
Auerswald H, Eng C, Lay S, In S, Eng S, Vo HTM, Sith C, Cheng S, Delvallez G, Mich V, Meng N, Sovann L, Sidonn K, Vanhomwegen J, Cantaert T, Dussart P, Duong V, Karlsson EA. Rapid Generation of In-House Serological Assays Is Comparable to Commercial Kits Critical for Early Response to Pandemics: A Case With SARS-CoV-2. Front Med (Lausanne) 2022; 9:864972. [PMID: 35602487 PMCID: PMC9121123 DOI: 10.3389/fmed.2022.864972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Accurate and sensitive measurement of antibodies is critical to assess the prevalence of infection, especially asymptomatic infection, and to analyze the immune response to vaccination during outbreaks and pandemics. A broad variety of commercial and in-house serological assays are available to cater to different laboratory requirements; however direct comparison is necessary to understand utility. Materials and Methods We investigate the performance of six serological methods against SARS-CoV-2 to determine the antibody profile of 250 serum samples, including 234 RT-PCR-confirmed SARS-CoV-2 cases, the majority with asymptomatic presentation (87.2%) at 1-51 days post laboratory diagnosis. First, we compare to the performance of two in-house antibody assays: (i) an in-house IgG ELISA, utilizing UV-inactivated virus, and (ii) a live-virus neutralization assay (PRNT) using the same Cambodian isolate as the ELISA. In-house assays are then compared to standardized commercial anti-SARS-CoV-2 electrochemiluminescence immunoassays (Elecsys ECLIAs, Roche Diagnostics; targeting anti-N and anti-S antibodies) along with a flow cytometry based assay (FACS) that measures IgM and IgG against spike (S) protein and a multiplex microsphere-based immunoassay (MIA) determining the antibodies against various spike and nucleoprotein (N) antigens of SARS-CoV-2 and other coronaviruses (SARS-CoV-1, MERS-CoV, hCoVs 229E, NL63, HKU1). Results Overall, specificity of assays was 100%, except for the anti-S IgM flow cytometry based assay (96.2%), and the in-house IgG ELISA (94.2%). Sensitivity ranged from 97.3% for the anti-S ECLIA down to 76.3% for the anti-S IgG flow cytometry based assay. PRNT and in-house IgG ELISA performed similarly well when compared to the commercial ECLIA: sensitivity of ELISA and PRNT was 94.7 and 91.1%, respectively, compared to S- and N-targeting ECLIA with 97.3 and 96.8%, respectively. The MIA revealed cross-reactivity of antibodies from SARS-CoV-2-infected patients to the nucleocapsid of SARS-CoV-1, and the spike S1 domain of HKU1. Conclusion In-house serological assays, especially ELISA and PRNT, perform similarly to commercial assays, a critical factor in pandemic response. Selection of suitable immunoassays should be made based on available resources and diagnostic needs.
Collapse
Affiliation(s)
- Heidi Auerswald
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Chanreaksmey Eng
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sokchea Lay
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Saraden In
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sokchea Eng
- Medical Biology Laboratory, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Hoa Thi My Vo
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Charya Sith
- Medical Biology Laboratory, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sokleaph Cheng
- Medical Biology Laboratory, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Gauthier Delvallez
- Medical Biology Laboratory, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Vann Mich
- Khmer–Soviet Friendship Hospital, Ministry of Health, Phnom Penh, Cambodia
| | - Ngy Meng
- Khmer–Soviet Friendship Hospital, Ministry of Health, Phnom Penh, Cambodia
| | - Ly Sovann
- Communicable Disease Control Department, Ministry of Health, Phnom Penh, Cambodia
| | - Kraing Sidonn
- Communicable Disease Control Department, Ministry of Health, Phnom Penh, Cambodia
| | | | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Philippe Dussart
- Institut Pasteur de Madagascar, Pasteur Network, Antananarivo, Madagascar
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Erik A. Karlsson
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| |
Collapse
|
21
|
Dewhurst RE, Heinrich T, Watt P, Ostergaard P, Marimon JM, Moreira M, Houldsworth PE, Rudrum JD, Wood D, Kõks S. Validation of a rapid, saliva-based, and ultra-sensitive SARS-CoV-2 screening system for pandemic-scale infection surveillance. Sci Rep 2022; 12:5936. [PMID: 35395856 PMCID: PMC8990279 DOI: 10.1038/s41598-022-08263-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/02/2022] [Indexed: 12/23/2022] Open
Abstract
Without any realistic prospect of comprehensive global vaccine coverage and lasting immunity, control of pandemics such as COVID-19 will require implementation of large-scale, rapid identification and isolation of infectious individuals to limit further transmission. Here, we describe an automated, high-throughput integrated screening platform, incorporating saliva-based loop-mediated isothermal amplification (LAMP) technology, that is designed for population-scale sensitive detection of infectious carriers of SARS-CoV-2 RNA. Central to this surveillance system is the "Sentinel" testing instrument, which is capable of reporting results within 25 min of saliva sample collection with a throughput of up to 3840 results per hour. It incorporates continuous flow loading of samples at random intervals to cost-effectively adjust for fluctuations in testing demand. Independent validation of our saliva-based RT-LAMP technology on an automated LAMP instrument coined the "Sentinel", found 98.7% sensitivity, 97.6% specificity, and 98% accuracy against a RT-PCR comparator assay, confirming its suitability for surveillance screening. This Sentinel surveillance system offers a feasible and scalable approach to complement vaccination, to curb the spread of COVID-19 variants, and control future pandemics to save lives.
Collapse
Affiliation(s)
- Robert E Dewhurst
- Perron Institute for Neurological and Translational Science, Perth, WA, 6009, Australia
- Avicena Systems Ltd, West Perth, WA, 6005, Australia
| | - Tatjana Heinrich
- Perron Institute for Neurological and Translational Science, Perth, WA, 6009, Australia
- Avicena Systems Ltd, West Perth, WA, 6005, Australia
| | - Paul Watt
- Avicena Systems Ltd, West Perth, WA, 6005, Australia
- Telethon Kids Institute, University of Western Australia, Perth, WA, 6009, Australia
| | | | - Jose M Marimon
- Biodonostia Health Research Institute, Infectious Diseases Area, Osakidetza Basque Health Service, Donostialdea Integrated Health Organization, San Sebastián, Spain
| | - Mariana Moreira
- Lancs Lamp Laboratory, Heatley House, Bowran Street, Preston, PR1 2UX, UK
| | | | - Jack D Rudrum
- Perron Institute for Neurological and Translational Science, Perth, WA, 6009, Australia
- Avicena Systems Ltd, West Perth, WA, 6005, Australia
| | - David Wood
- University of Western Australia, Perth, WA, 6009, Australia
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, Perth, WA, 6009, Australia.
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, 6150, Australia.
- Prion Ltd, 50410, Tartu, Estonia.
| |
Collapse
|
22
|
Vindeirinho JM, Pinho E, Azevedo NF, Almeida C. SARS-CoV-2 Diagnostics Based on Nucleic Acids Amplification: From Fundamental Concepts to Applications and Beyond. Front Cell Infect Microbiol 2022; 12:799678. [PMID: 35402302 PMCID: PMC8984495 DOI: 10.3389/fcimb.2022.799678] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
COVID-19 pandemic ignited the development of countless molecular methods for the diagnosis of SARS-CoV-2 based either on nucleic acid, or protein analysis, with the first establishing as the most used for routine diagnosis. The methods trusted for day to day analysis of nucleic acids rely on amplification, in order to enable specific SARS-CoV-2 RNA detection. This review aims to compile the state-of-the-art in the field of nucleic acid amplification tests (NAATs) used for SARS-CoV-2 detection, either at the clinic level, or at the Point-Of-Care (POC), thus focusing on isothermal and non-isothermal amplification-based diagnostics, while looking carefully at the concerning virology aspects, steps and instruments a test can involve. Following a theme contextualization in introduction, topics about fundamental knowledge on underlying virology aspects, collection and processing of clinical samples pave the way for a detailed assessment of the amplification and detection technologies. In order to address such themes, nucleic acid amplification methods, the different types of molecular reactions used for DNA detection, as well as the instruments requested for executing such routes of analysis are discussed in the subsequent sections. The benchmark of paradigmatic commercial tests further contributes toward discussion, building on technical aspects addressed in the previous sections and other additional information supplied in that part. The last lines are reserved for looking ahead to the future of NAATs and its importance in tackling this pandemic and other identical upcoming challenges.
Collapse
Affiliation(s)
- João M. Vindeirinho
- National Institute for Agrarian and Veterinarian Research (INIAV, I.P), Vairão, Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, Porto, Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering, University of Porto, Porto, Portugal
| | - Eva Pinho
- National Institute for Agrarian and Veterinarian Research (INIAV, I.P), Vairão, Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, Porto, Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering, University of Porto, Porto, Portugal
| | - Nuno F. Azevedo
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, Porto, Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering, University of Porto, Porto, Portugal
| | - Carina Almeida
- National Institute for Agrarian and Veterinarian Research (INIAV, I.P), Vairão, Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, Porto, Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering, University of Porto, Porto, Portugal
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| |
Collapse
|
23
|
Vo HTM, Maestri A, Auerswald H, Sorn S, Lay S, Seng H, Sann S, Ya N, Pean P, Dussart P, Schwartz O, Ly S, Bruel T, Ly S, Duong V, Karlsson EA, Cantaert T. Robust and Functional Immune Memory Up to 9 Months After SARS-CoV-2 Infection: A Southeast Asian Longitudinal Cohort. Front Immunol 2022; 13:817905. [PMID: 35185909 PMCID: PMC8853741 DOI: 10.3389/fimmu.2022.817905] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/10/2022] [Indexed: 01/14/2023] Open
Abstract
The duration of humoral and cellular immune memory following SARS-CoV-2 infection in populations in least developed countries remains understudied but is key to overcome the current SARS-CoV-2 pandemic. Sixty-four Cambodian individuals with laboratory-confirmed infection with asymptomatic or mild/moderate clinical presentation were evaluated for Spike (S)-binding and neutralizing antibodies and antibody effector functions during acute phase of infection and at 6-9 months follow-up. Antigen-specific B cells, CD4+ and CD8+ T cells were characterized, and T cells were interrogated for functionality at late convalescence. Anti-S antibody titers decreased over time, but effector functions mediated by S-specific antibodies remained stable. S- and nucleocapsid (N)-specific B cells could be detected in late convalescence in the activated memory B cell compartment and are mostly IgG+. CD4+ and CD8+ T cell immune memory was maintained to S and membrane (M) protein. Asymptomatic infection resulted in decreased antibody-dependent cellular cytotoxicity (ADCC) and frequency of SARS-CoV-2-specific CD4+ T cells at late convalescence. Whereas anti-S antibodies correlated with S-specific B cells, there was no correlation between T cell response and humoral immune memory. Hence, all aspects of a protective immune response are maintained up to nine months after SARS-CoV-2 infection and in the absence of re-infection.
Collapse
Affiliation(s)
- Hoa Thi My Vo
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Alvino Maestri
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Heidi Auerswald
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sopheak Sorn
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sokchea Lay
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Heng Seng
- Department of Communicable Disease Control, Ministry of Health (CDC-MoH), Phnom Penh, Cambodia
| | - Sotheary Sann
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Nisa Ya
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Polidy Pean
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Philippe Dussart
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Olivier Schwartz
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France.,Vaccine Research Institute, Créteil, France
| | - Sovann Ly
- Department of Communicable Disease Control, Ministry of Health (CDC-MoH), Phnom Penh, Cambodia
| | - Timothée Bruel
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France.,Vaccine Research Institute, Créteil, France
| | - Sowath Ly
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Erik A Karlsson
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| |
Collapse
|
24
|
Zhang D, Duran SSF, Lim WYS, Tan CKI, Cheong WCD, Suwardi A, Loh XJ. SARS-CoV-2 in wastewater: From detection to evaluation. MATERIALS TODAY. ADVANCES 2022; 13:100211. [PMID: 35098102 PMCID: PMC8786653 DOI: 10.1016/j.mtadv.2022.100211] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/16/2022] [Accepted: 01/21/2022] [Indexed: 05/07/2023]
Abstract
SARS-CoV-2 presence in wastewater has been reported in several studies and has received widespread attention among the Wastewater-based epidemiology (WBE) community. Such studies can potentially be used as a proxy for early warning of potential COVID-19 outbreak, or as a mitigation measure for potential virus transmission via contaminated water. In this review, we summarized the latest understanding on the detection, concentration, and evaluation of SARS-CoV-2 in wastewater. Importantly, we discuss factors affecting the quality of wastewater surveillance ranging from temperature, pH, starting concentration, as well as the presence of chemical pollutants. These factors greatly affect the reliability and comparability of studies reported by various communities across the world. Overall, this review provides a broadly encompassing guidance for epidemiological study using wastewater surveillance.
Collapse
Affiliation(s)
- Danwei Zhang
- Institute of Materials Research and Engineering, 2 Fusionopolis Way, Agency for Science, Technology and Research, Singapore, 138634
| | - Solco S Faye Duran
- Institute of Materials Research and Engineering, 2 Fusionopolis Way, Agency for Science, Technology and Research, Singapore, 138634
| | - Wei Yang Samuel Lim
- Institute of Materials Research and Engineering, 2 Fusionopolis Way, Agency for Science, Technology and Research, Singapore, 138634
| | - Chee Kiang Ivan Tan
- Institute of Materials Research and Engineering, 2 Fusionopolis Way, Agency for Science, Technology and Research, Singapore, 138634
| | - Wun Chet Davy Cheong
- Institute of Materials Research and Engineering, 2 Fusionopolis Way, Agency for Science, Technology and Research, Singapore, 138634
| | - Ady Suwardi
- Institute of Materials Research and Engineering, 2 Fusionopolis Way, Agency for Science, Technology and Research, Singapore, 138634
| | - Xian Jun Loh
- Institute of Materials Research and Engineering, 2 Fusionopolis Way, Agency for Science, Technology and Research, Singapore, 138634
| |
Collapse
|
25
|
Woldesemayat B, Gebremicael G, Zealiyas K, Yilma A, Adane S, Yimer M, Gutema G, Feleke A, Desta K. Effect of heat inactivation for the detection of severe acute respiratory syndrome-corona virus-2 (SARS-CoV-2) with reverse transcription real time polymerase chain reaction (rRT-PCR): evidence from Ethiopian study. BMC Infect Dis 2022; 22:163. [PMID: 35189815 PMCID: PMC8860295 DOI: 10.1186/s12879-022-07134-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) has been a major public health importance and its specimen needs to be handled safely due to concerns of potential transmissibility to health care workers. Heat inactivation of the sample before nucleic acid isolation might permit safe testing processes. Hence, it is important to assess the effect of heat inactivation on SARS-CoV-2 RT-PCR detection in resource limited settings. Methods An experimental study was conducted at Ethiopian Public Health Institute (EPHI) from September 25 to October 15, 2020. A total of 188 Oro-pharyngeal swabs were collected from COVID-19 suspected cases, referred to EPHI for SARS COV-2 testing. One batch of the sample was inactivated at 56 °C heat for 30 min, and the other batch was stored at 4 °C for a similar period of time. RNA extraction and detection were done by DAAN Gene kit protocols. Abbott m2000 RT-PCR was used for amplification and detection. Data analysis was done by using SPSS version 23.0; Chi-square and Pearson correlation test for qualitative and semi-quantitative analysis were used. p-value < 0.05 was considered as statistically significant. Results Out of 188 total samples, 119 (63.3%) were positive and 69 (36.7%) were negative in the non-inactivated group. While, 115 (61.2%) of samples were positive and 73 (38.8) were negative in heat inactivated sample batch. Rate of positivity between groups did not have statistically significant difference (p > 0.05). The mean Cycle threshold (Ct) value difference between the two groups of ORF1a/b gene and N gene was 0.042 (95% CI − 0.247–0.331; t = 0.28; p = 0.774) and 0.38 (95% CI 0.097–0.682; t = 2.638; p = 0.010) respectively. Conclusion Heat inactivation at 56 °C for 30 min did not affect the qualitative rRT-PCR detection of SARS-CoV-2. However, the finding showed that there was statistically significant Ct value increment after heat inactivation compared to untreated samples. Therefore, false-negative results for high Ct value (Ct > 35) samples were found to be the challenge of this protocol. Hence alternative inactivation methods should be investigated and further studies should be considered.
Collapse
Affiliation(s)
- Belete Woldesemayat
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, P.O. Box 1242, Addis Ababa, Ethiopia.
| | - Gebremedihin Gebremicael
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, P.O. Box 1242, Addis Ababa, Ethiopia
| | - Kidist Zealiyas
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, P.O. Box 1242, Addis Ababa, Ethiopia
| | - Amelework Yilma
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, P.O. Box 1242, Addis Ababa, Ethiopia
| | - Sisay Adane
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, P.O. Box 1242, Addis Ababa, Ethiopia
| | - Mengistu Yimer
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, P.O. Box 1242, Addis Ababa, Ethiopia
| | - Gadissa Gutema
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, P.O. Box 1242, Addis Ababa, Ethiopia
| | - Altaye Feleke
- HIV/AIDS Disease Research Team, TB and HIV/AIDS Disease Research Directorate, Ethiopian Public Health Institute, P.O. Box 1242, Addis Ababa, Ethiopia
| | - Kassu Desta
- Department of Medical Laboratory Sciences, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
26
|
Delpuech O, Douthwaite JA, Hill T, Niranjan D, Malintan NT, Duvoisin H, Elliott J, Goodfellow I, Hosmillo M, Orton AL, Taylor MA, Brankin C, Pitt H, Ross-Thriepland D, Siek M, Cuthbert A, Richards I, Ferdinand JR, Barker C, Shaw R, Ariani C, Waddell I, Rees S, Green C, Clark R, Upadhyay A, Howes R. Heat inactivation of clinical COVID-19 samples on an industrial scale for low risk and efficient high-throughput qRT-PCR diagnostic testing. Sci Rep 2022; 12:2883. [PMID: 35190592 PMCID: PMC8861189 DOI: 10.1038/s41598-022-06888-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 02/03/2022] [Indexed: 12/28/2022] Open
Abstract
We report the development of a large scale process for heat inactivation of clinical COVID-19 samples prior to laboratory processing for detection of SARS-CoV-2 by RT-qPCR. With more than 266 million confirmed cases, over 5.26 million deaths already recorded at the time of writing, COVID-19 continues to spread in many parts of the world. Consequently, mass testing for SARS-CoV-2 will remain at the forefront of the COVID-19 response and prevention for the near future. Due to biosafety considerations the standard testing process requires a significant amount of manual handling of patient samples within calibrated microbiological safety cabinets. This makes the process expensive, effects operator ergonomics and restricts testing to higher containment level laboratories. We have successfully modified the process by using industrial catering ovens for bulk heat inactivation of oropharyngeal/nasopharyngeal swab samples within their secondary containment packaging before processing in the lab to enable all subsequent activities to be performed in the open laboratory. As part of a validation process, we tested greater than 1200 clinical COVID-19 samples and showed less than 1 Cq loss in RT-qPCR test sensitivity. We also demonstrate the bulk heat inactivation protocol inactivates a murine surrogate of human SARS-CoV-2. Using bulk heat inactivation, the assay is no longer reliant on containment level 2 facilities and practices, which reduces cost, improves operator safety and ergonomics and makes the process scalable. In addition, heating as the sole method of virus inactivation is ideally suited to streamlined and more rapid workflows such as 'direct to PCR' assays that do not involve RNA extraction or chemical neutralisation methods.
Collapse
Affiliation(s)
- Oona Delpuech
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Julie A Douthwaite
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK.
- In Vivo Expressed Biologics, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK.
| | - Thomas Hill
- Charles River Laboratories, Chesterford Research Park, Saffron Walden, CB10 1XL, UK
| | - Dhevahi Niranjan
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Nancy T Malintan
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Hannah Duvoisin
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Jane Elliott
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Ian Goodfellow
- Division of Virology, Department of Pathology, Addenbrooke's Hospital, Cambridge, UK
| | - Myra Hosmillo
- Division of Virology, Department of Pathology, Addenbrooke's Hospital, Cambridge, UK
| | - Alexandra L Orton
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Molly A Taylor
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Christopher Brankin
- Biologics Engineering, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Haidee Pitt
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
- Animal Science and Technologies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Magdalena Siek
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
- Facilities Management, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Anna Cuthbert
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
- Clinical Operations, Late-Stage Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Ian Richards
- Charles River Laboratories, Chesterford Research Park, Saffron Walden, CB10 1XL, UK
| | - John R Ferdinand
- Charles River Laboratories, Chesterford Research Park, Saffron Walden, CB10 1XL, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Robert Shaw
- Oral Product Development, Pharmaceutical Technology and Development, Operations, AstraZeneca, Macclesfield, UK
| | | | - Ian Waddell
- Charles River Laboratories, Chesterford Research Park, Saffron Walden, CB10 1XL, UK
| | - Steve Rees
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Clive Green
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Roger Clark
- Charles River Laboratories, Chesterford Research Park, Saffron Walden, CB10 1XL, UK
| | | | - Rob Howes
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
27
|
Jia M, Taylor TM, Senger SM, Ovissipour R, Bertke AS. SARS-CoV-2 Remains Infectious on Refrigerated Deli Food, Meats, and Fresh Produce for up to 21 Days. Foods 2022; 11:286. [PMID: 35159438 PMCID: PMC8834215 DOI: 10.3390/foods11030286] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2, the virus that causes COVID-19, has been detected on foods and food packaging and the virus can infect oral cavity and intestinal cells, suggesting that infection could potentially occur following ingestion of virus-contaminated foods. To determine the relative risk of infection from different types of foods, we assessed survival of SARS-CoV-2 on refrigerated ready-to-eat deli items, fresh produce, and meats (including seafood). Deli items and meats with high protein, fat, and moisture maintained infectivity of SARS-CoV-2 for up to 21 days. However, processed meat, such as salami, and some fresh produce exhibited antiviral effects. SARS-CoV-2 also remained infectious in ground beef cooked rare or medium, but not well-done. Although infectious SARS-CoV-2 was inactivated on the foods over time, viral RNA was not degraded in similar trends, regardless of food type; thus, PCR-based assays for detection of pathogens on foods only indicate the presence of viral RNA, but do not correlate with presence or quantity of infectious virus. The survival and high recovery of SARS-CoV-2 on certain foods support the possibility that food contaminated with SARS-CoV-2 could potentially be a source of infection, highlighting the importance of proper food handling and cooking to inactivate any contaminating virus prior to consumption.
Collapse
Affiliation(s)
- Mo Jia
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; (M.J.); (T.M.T.)
| | - Tina M. Taylor
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; (M.J.); (T.M.T.)
| | - Sterling M. Senger
- Food Science and Technology, Agricultural Research and Extension Center, Virginia Polytechnic Institute & State University, Hampton, VA 23669, USA; (S.M.S.); (R.O.)
| | - Reza Ovissipour
- Food Science and Technology, Agricultural Research and Extension Center, Virginia Polytechnic Institute & State University, Hampton, VA 23669, USA; (S.M.S.); (R.O.)
- Center for Emerging Zoonotic and Arthropod-Borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Andrea S. Bertke
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; (M.J.); (T.M.T.)
- Center for Emerging Zoonotic and Arthropod-Borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| |
Collapse
|
28
|
Wu F, Xiao A, Zhang J, Moniz K, Endo N, Armas F, Bonneau R, Brown MA, Bushman M, Chai PR, Duvallet C, Erickson TB, Foppe K, Ghaeli N, Gu X, Hanage WP, Huang KH, Lee WL, Matus M, McElroy KA, Nagler J, Rhode SF, Santillana M, Tucker JA, Wuertz S, Zhao S, Thompson J, Alm EJ. SARS-CoV-2 RNA concentrations in wastewater foreshadow dynamics and clinical presentation of new COVID-19 cases. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 805:150121. [PMID: 34534872 PMCID: PMC8416286 DOI: 10.1016/j.scitotenv.2021.150121] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 05/18/2023]
Abstract
Current estimates of COVID-19 prevalence are largely based on symptomatic, clinically diagnosed cases. The existence of a large number of undiagnosed infections hampers population-wide investigation of viral circulation. Here, we quantify the SARS-CoV-2 concentration and track its dynamics in wastewater at a major urban wastewater treatment facility in Massachusetts, between early January and May 2020. SARS-CoV-2 was first detected in wastewater on March 3. SARS-CoV-2 RNA concentrations in wastewater correlated with clinically diagnosed new COVID-19 cases, with the trends appearing 4-10 days earlier in wastewater than in clinical data. We inferred viral shedding dynamics by modeling wastewater viral load as a convolution of back-dated new clinical cases with the average population-level viral shedding function. The inferred viral shedding function showed an early peak, likely before symptom onset and clinical diagnosis, consistent with emerging clinical and experimental evidence. This finding suggests that SARS-CoV-2 concentrations in wastewater may be primarily driven by viral shedding early in infection. This work shows that longitudinal wastewater analysis can be used to identify trends in disease transmission in advance of clinical case reporting, and infer early viral shedding dynamics for newly infected individuals, which are difficult to capture in clinical investigations.
Collapse
Affiliation(s)
- Fuqing Wu
- Department of Biological Engineering, Massachusetts Institute of Technology, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, USA
| | - Amy Xiao
- Department of Biological Engineering, Massachusetts Institute of Technology, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, USA
| | - Jianbo Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, USA
| | - Katya Moniz
- Department of Biological Engineering, Massachusetts Institute of Technology, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, USA
| | | | - Federica Armas
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance Interdisciplinary Research Group, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | - Richard Bonneau
- Center for Data Science NYU, Center for Social Media and Politics, New York University, USA
| | - Megan A Brown
- Center for Data Science NYU, Center for Social Media and Politics, New York University, USA
| | - Mary Bushman
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Peter R Chai
- Division of Medical Toxicology, Department of Emergency Medicine, Brigham and Women's Hospital, Harvard Medical School, USA; The Fenway Institute, Fenway Health, Boston, MA, USA
| | | | - Timothy B Erickson
- Division of Medical Toxicology, Department of Emergency Medicine, Brigham and Women's Hospital, Harvard Medical School, USA; Harvard Humanitarian Initiative, Harvard University, USA
| | | | | | - Xiaoqiong Gu
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance Interdisciplinary Research Group, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | | | | | - Wei Lin Lee
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance Interdisciplinary Research Group, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | | | | | - Jonathan Nagler
- Center for Data Science NYU, Center for Social Media and Politics, New York University, USA
| | - Steven F Rhode
- Massachusetts Water Resources Authority, Boston, MA, USA
| | - Mauricio Santillana
- Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA; Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
| | - Joshua A Tucker
- Center for Data Science NYU, Center for Social Media and Politics, New York University, USA
| | - Stefan Wuertz
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore; School of Civil and Environmental Enginering, Nanyang Technological University, Singapore
| | - Shijie Zhao
- Department of Biological Engineering, Massachusetts Institute of Technology, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, USA
| | - Janelle Thompson
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore; Asian School of the Environment, Nanyang Technological University, Singapore
| | - Eric J Alm
- Department of Biological Engineering, Massachusetts Institute of Technology, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, USA; Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance Interdisciplinary Research Group, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
29
|
Canpolat M, Bozkurt S, Şakalar Ç, Çoban AY, Karaçaylı D, Toker E. Rapid thermal inactivation of aerosolized SARS-CoV-2. J Virol Methods 2022; 301:114465. [PMID: 35033579 PMCID: PMC8757645 DOI: 10.1016/j.jviromet.2022.114465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/27/2021] [Accepted: 01/11/2022] [Indexed: 12/04/2022]
Abstract
Airborne transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is one of the leading mechanisms of spread, especially in confined environments. The study aims to assess the thermal inactivation of SARS-CoV-2 at high temperatures in the time scale of seconds. An electric heater with a coiled resistance wire is located perpendicularly to the airflow direction inside an air tunnel. The airflow rate through the tunnel was 0.6 m3/h (10 L/ min). SARS-CoV-2 were suspended in Dulbecco’s modified Eagle’s medium (DMEM) with 10 % fetal bovine serum (FBS), aerosolized by a nebulizer at a rate of 0.2 L/min and introduced to the airflow inside the heater with the use of a compressor and an aspirator. In the control experiment, with the heater off, SARS-CoV-2 passed through the system. In the virus inactivation test experiments, the heater’s outlet air temperature was set to 150 ± 5 °C and 220 ± 5 °C, and the air traveling through the tunnel was exposed to heat for 1.44 s. An inline gelatine filter harvested SARS-CoV-2 that passed through the system. The viral titer obtained from the gelatine filter in the control experiment was about 5.5 log10 TCID50. The virus's loss in viability in test experiments at 150 °C and 220 °C were 99.900 % and 99.999 %, respectively. The results indicate that high-temperature thermal inactivation substantially reduces the concentration of SARS-CoV-2 in the air within seconds.
Collapse
Affiliation(s)
- Murat Canpolat
- Biomedical Optics Research Unit, Department of Biophysics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey.
| | - Serhat Bozkurt
- Department of Gerontology, Faculty of Health Sciences, Akdeniz University, Antalya 07070, Turkey
| | - Çağrı Şakalar
- Antimikrop Ar-Ge ve Biyosidal Analiz Merkezi, Nasuh Akar Mah, Süleyman Hacıabdullahoğlu Cad. No: 37/1, Çankaya, Ankara, Turkey
| | - Ahmet Yılmaz Çoban
- Tuberculosis Research Center, Akdeniz University, Antalya 07070, Turkey; Department of Nutrition & Dietetics, Faculty of Health Sciences, Akdeniz University, Antalya 07070, Turkey
| | - Deniz Karaçaylı
- Biomedical Optics Research Unit, Department of Biophysics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Emre Toker
- College of Agriculture & Life Sciences, University of Arizona, Saguaro Hall 129, 110 E. South Campus Dr. Tucson Arizona, AZ 87571-0033, USA
| |
Collapse
|
30
|
Alterations of fecal antibiotic resistome in COVID-19 patients after empirical antibiotic exposure. Int J Hyg Environ Health 2021; 240:113882. [PMID: 34915282 PMCID: PMC8664710 DOI: 10.1016/j.ijheh.2021.113882] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 11/05/2021] [Accepted: 11/16/2021] [Indexed: 12/16/2022]
Abstract
As the COVID-19 pandemic spread globally, the consumption of antibiotics increased. However, no studies exist evaluating the effect of antibiotics use on the antibiotic resistance of intestinal flora in COVID-19 patients during the pandemic. To explore this issue, we collected 15 metagenomic data of fecal samples from healthy controls (HCs) with no use history of antibiotics, 23 metagenomic data of fecal samples from COVID-19 patients who received empirical antibiotics [COVID-19 (abx+)], 18 metagenomic data of fecal samples from antibiotics-naïve COVID-19 patients [COVID-19 (abx-)], and six metagenomic data of fecal samples from patients with community-acquired pneumonia [PC (abx+)] from the Sequence Read Archive database. A total of 513 antibiotic-resistant gene (ARG) subtypes of 18 ARG types were found. Antibiotic treatment resulted in a significant increase in the abundance of ARGs in intestinal flora of COVID-19 patients and markedly altered the composition of ARG profiles. Grouped comparisons of pairs of Bray-Curtis dissimilarity values demonstrated that the dissimilarity of the HC versus the COVID-19 (abx+) group was significantly higher than the dissimilarity of the HC versus the COVID-19 (abx-) group. The mexF, mexD, OXA_209, major facilitator superfamily transporter, and EmrB_QacA family major facilitator transporter genes were the discriminative ARG subtypes for the COVID-19 (abx+) group. IS621, qacEdelta, transposase, and ISCR were significantly increased in COVID-19 (abx+) group; they greatly contributed toward explaining variation in the relative abundance of ARG types. Overall, our data provide important insights into the effect of antibiotics use on the antibiotic resistance of COVID-19 patients during the COVID-19 epidemic.
Collapse
|
31
|
Andriamandimby SF, Brook CE, Razanajatovo N, Randriambolamanantsoa TH, Rakotondramanga JM, Rasambainarivo F, Raharimanga V, Razanajatovo IM, Mangahasimbola R, Razafindratsimandresy R, Randrianarisoa S, Bernardson B, Rabarison JH, Randrianarisoa M, Nasolo FS, Rabetombosoa RM, Ratsimbazafy AM, Raharinosy V, Rabemananjara AH, Ranaivoson CH, Razafimanjato H, Randremanana R, Héraud JM, Dussart P. Cross-sectional cycle threshold values reflect epidemic dynamics of COVID-19 in Madagascar. Epidemics 2021; 38:100533. [PMID: 34896895 PMCID: PMC8628610 DOI: 10.1016/j.epidem.2021.100533] [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: 06/09/2021] [Revised: 09/23/2021] [Accepted: 11/27/2021] [Indexed: 01/12/2023] Open
Abstract
As the national reference laboratory for febrile illness in Madagascar, we processed samples from the first epidemic wave of COVID-19, between March and September 2020. We fit generalized additive models to cycle threshold (Ct) value data from our RT-qPCR platform, demonstrating a peak in high viral load, low-Ct value infections temporally coincident with peak epidemic growth rates estimated in real time from publicly-reported incidence data and retrospectively from our own laboratory testing data across three administrative regions. We additionally demonstrate a statistically significant effect of duration of time since infection onset on Ct value, suggesting that Ct value can be used as a biomarker of the stage at which an individual is sampled in the course of an infection trajectory. As an extension, the population-level Ct distribution at a given timepoint can be used to estimate population-level epidemiological dynamics. We illustrate this concept by adopting a recently-developed, nested modeling approach, embedding a within-host viral kinetics model within a population-level Susceptible-Exposed-Infectious-Recovered (SEIR) framework, to mechanistically estimate epidemic growth rates from cross-sectional Ct distributions across three regions in Madagascar. We find that Ct-derived epidemic growth estimates slightly precede those derived from incidence data across the first epidemic wave, suggesting delays in surveillance and case reporting. Our findings indicate that public reporting of Ct values could offer an important resource for epidemiological inference in low surveillance settings, enabling forecasts of impending incidence peaks in regions with limited case reporting.
Collapse
Affiliation(s)
| | - Cara E Brook
- Department of Ecology and Evolution, University of Chicago, United States
| | | | | | | | | | | | | | | | | | | | - Barivola Bernardson
- Epidemiology and Clinical Research Unit, Institut Pasteur de Madagascar, Madagascar
| | | | | | | | | | | | | | | | | | | | - Rindra Randremanana
- Virology Unit, Institut Pasteur de Madagascar, Madagascar; Epidemiology and Clinical Research Unit, Institut Pasteur de Madagascar, Madagascar
| | | | | |
Collapse
|
32
|
Rowan NJ, Meade E, Garvey M. Efficacy of frontline chemical biocides and disinfection approaches for inactivating SARS-CoV-2 variants of concern that cause coronavirus disease with the emergence of opportunities for green eco-solutions. CURRENT OPINION IN ENVIRONMENTAL SCIENCE & HEALTH 2021; 23:100290. [PMID: 34250323 PMCID: PMC8254398 DOI: 10.1016/j.coesh.2021.100290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The emergence of severe acute respiratory disease (SARS-CoV-2) variants that cause coronavirus disease is of global concern. Severe acute respiratory disease variants of concern (VOC) exhibiting greater transmissibility, and potentially increased risk of hospitalization, severity and mortality, are attributed to molecular mutations in outer viral surface spike proteins. Thus, there is a reliance on using appropriate counter-disease measures, including non-pharmaceutical interventions and vaccination. The best evidence suggests that the use of frontline biocides effectively inactivate coronavirus similarly, including VOC, such as 202012/01, 501Y.V2 and P.1 that have rapidly replaced the wild-type variant in the United Kingdom, South Africa and Brazil, respectively. However, this review highlights that efficacy of VOC-disinfection will depend on the type of biocide and the parameters governing the activity. VOC are likely to be similar in size to the wild-type strain, thus implying that existing guidelines for use and re-use of face masks post disinfection remain relevant. Monitoring to avoid injudicious use of biocides during the coronavirus disease era is required as prolonged and excessive biocide usage may negatively impact our receiving environments; thus, highlighting the potential for alternative more environmental-friendly sustainable biocide solutions. Traditional biocides may promote cross-antimicrobial resistance to antibiotics in problematical bacteria. The existing filtration efficacy of face masks is likely to perform similarly for VOC due to similar viral size; however, advances in face mask manufacturing by way incorporating new anti-viral materials will potentially enhance their design and functionality for existing and potential future pandemics.
Collapse
Affiliation(s)
- Neil J Rowan
- Centre for Disinfection and Sterilisation, Athlone Institute of Technology, Dublin Road, Athlone, Ireland
- Department of Nursing and Healthcare, Athlone Institute of Technology, Dublin Road, Athlone, Ireland
| | - Elaine Meade
- Department of Life Science, Institute of Technology, Sligo, Ash Lane, Sligo, Ireland
| | - Mary Garvey
- Department of Life Science, Institute of Technology, Sligo, Ash Lane, Sligo, Ireland
| |
Collapse
|
33
|
Lista MJ, Matos PM, Maguire TJA, Poulton K, Ortiz-Zapater E, Page R, Sertkaya H, Ortega-Prieto AM, Scourfield E, O’Byrne AM, Bouton C, Dickenson RE, Ficarelli M, Jimenez-Guardeño JM, Howard M, Betancor G, Galao RP, Pickering S, Signell AW, Wilson H, Cliff P, Kia Ik MT, Patel A, MacMahon E, Cunningham E, Doores K, Agromayor M, Martin-Serrano J, Perucha E, Mischo HE, Shankar-Hari M, Batra R, Edgeworth J, Zuckerman M, Malim MH, Neil S, Martinez-Nunez RT. Resilient SARS-CoV-2 diagnostics workflows including viral heat inactivation. PLoS One 2021; 16:e0256813. [PMID: 34525109 PMCID: PMC8443028 DOI: 10.1371/journal.pone.0256813] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/17/2021] [Indexed: 12/23/2022] Open
Abstract
There is a worldwide need for reagents to perform SARS-CoV-2 detection. Some laboratories have implemented kit-free protocols, but many others do not have the capacity to develop these and/or perform manual processing. We provide multiple workflows for SARS-CoV-2 nucleic acid detection in clinical samples by comparing several commercially available RNA extraction methods: QIAamp Viral RNA Mini Kit (QIAgen), RNAdvance Blood/Viral (Beckman) and Mag-Bind Viral DNA/RNA 96 Kit (Omega Bio-tek). We also compared One-step RT-qPCR reagents: TaqMan Fast Virus 1-Step Master Mix (FastVirus, ThermoFisher Scientific), qPCRBIO Probe 1-Step Go Lo-ROX (PCR Biosystems) and Luna® Universal Probe One-Step RT-qPCR Kit (Luna, NEB). We used primer-probes that detect viral N (EUA CDC) and RdRP. RNA extraction methods provided similar results, with Beckman performing better with our primer-probe combinations. Luna proved most sensitive although overall the three reagents did not show significant differences. N detection was more reliable than that of RdRP, particularly in samples with low viral titres. Importantly, we demonstrated that heat treatment of nasopharyngeal swabs at 70°C for 10 or 30 min, or 90°C for 10 or 30 min (both original variant and B 1.1.7) inactivated SARS-CoV-2 employing plaque assays, and had minimal impact on the sensitivity of the qPCR in clinical samples. These findings make SARS-CoV-2 testing portable in settings that do not have CL-3 facilities. In summary, we provide several testing pipelines that can be easily implemented in other laboratories and have made all our protocols and SOPs freely available at https://osf.io/uebvj/.
Collapse
Affiliation(s)
- Maria Jose Lista
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Pedro M. Matos
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Thomas J. A. Maguire
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Inflammation Biology, School of Immunology and Microbial Sciences, Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, London, United Kingdom
| | - Kate Poulton
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Elena Ortiz-Zapater
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Randall Centre for Cell & Molecular Biophysics, King’s College London, London, United Kingdom
- Peter Gorer Department of Immunobiology, King’s College London, London, United Kingdom
| | - Robert Page
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- King’s Health Partners Integrated Cancer Centre, School of Cancer and Pharmaceutical Sciences, Guy’s Hospital, King’s College London, London, United Kingdom
| | - Helin Sertkaya
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Ana M. Ortega-Prieto
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Edward Scourfield
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Aoife M. O’Byrne
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Centre for Rheumatic Diseases (CRD–EULAR Centre of Excellence), King’s College London, London, United Kingdom
| | - Clement Bouton
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Ruth E. Dickenson
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Mattia Ficarelli
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Jose M. Jimenez-Guardeño
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Mark Howard
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Peter Gorer Department of Immunobiology, King’s College London, London, United Kingdom
| | - Gilberto Betancor
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Rui Pedro Galao
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Suzanne Pickering
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Adrian W. Signell
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Harry Wilson
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Penelope Cliff
- Viapath pathology laboratories at St Thomas’ Hospital, London, United Kingdom
| | - Mark Tan Kia Ik
- Centre for Infectious Diseases Research, St Thomas’ Hospital, London, United Kingdom
| | - Amita Patel
- Centre for Infectious Diseases Research, St Thomas’ Hospital, London, United Kingdom
| | - Eithne MacMahon
- Centre for Infectious Diseases Research, St Thomas’ Hospital, London, United Kingdom
| | - Emma Cunningham
- Centre for Infectious Diseases Research, St Thomas’ Hospital, London, United Kingdom
| | - Katie Doores
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Monica Agromayor
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Juan Martin-Serrano
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Esperanza Perucha
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Centre for Rheumatic Diseases (CRD–EULAR Centre of Excellence), King’s College London, London, United Kingdom
| | - Hannah E. Mischo
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Manu Shankar-Hari
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Rahul Batra
- Centre for Infectious Diseases Research, St Thomas’ Hospital, London, United Kingdom
| | - Jonathan Edgeworth
- Centre for Infectious Diseases Research, St Thomas’ Hospital, London, United Kingdom
| | - Mark Zuckerman
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- South London Specialist Virology Centre, King’s College Hospital, London, United Kingdom
| | - Michael H. Malim
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Stuart Neil
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Rocio Teresa Martinez-Nunez
- King’s College London Diagnostics Team at Guy’s Campus, London, United Kingdom
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
34
|
Taher J, Randell EW, Arnoldo S, Bailey D, De Guire V, Kaur S, Knauer M, Petryayeva E, Poutanen SM, Shaw JLV, Uddayasankar U, White-Al Habeeb N, Konforte D. Canadian Society of Clinical Chemists (CSCC) consensus guidance for testing, selection and quality management of SARS-CoV-2 point-of-care tests. Clin Biochem 2021; 95:1-12. [PMID: 34048776 PMCID: PMC8144094 DOI: 10.1016/j.clinbiochem.2021.05.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/02/2021] [Accepted: 05/22/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVES A consensus guidance is provided for testing, utility and verification of SARS-CoV-2 point-of-care test (POCT) performance and implementation of a quality management program, focusing on nucleic acid and antigen targeted technologies. DESIGN AND METHODS The recommendations are based on current literature and expert opinion from the members of Canadian Society of Clinical Chemists (CSCC), and are intended for use inside or outside of healthcare settings that have varied levels of expertise and experience with POCT. RESULTS AND CONCLUSIONS Here we discuss sampling requirements, biosafety, SARS-CoV-2 point-of-care testing methodologies (with focus on Health Canada approved tests), test performance and limitations, test selection, testing utility, development and implementation of quality management systems, quality improvement, and medical and scientific oversight.
Collapse
Affiliation(s)
- Jennifer Taher
- Pathology and Laboratory Medicine, Sinai Health System, Toronto, Canada; University of Toronto, Laboratory Medicine and Pathobiology, Toronto, Canada
| | - Edward W Randell
- Department of Laboratory Medicine, Faculty of Medicine, Memorial University of Newfoundland, Newfoundland, Canada
| | - Saranya Arnoldo
- University of Toronto, Laboratory Medicine and Pathobiology, Toronto, Canada; William Osler Health System, Brampton, Canada
| | | | - Vincent De Guire
- Clinical Biochemistry, Maisonneuve-Rosemont Hospital, Optilab-CHUM Laboratory Network, Montreal, Canada; Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Canada
| | - Sukhbir Kaur
- Fraser Health Authority, Vancouver, Canada; Pathology and Laboratory Medicine, University of British Columbia, Canada
| | - Michael Knauer
- Pathology and Laboratory Medicine, London Health Sciences Center, London, Canada; Pathology and Laboratory Medicine, University of Western Ontario, London, Canada
| | - Eleonora Petryayeva
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Susan M Poutanen
- University of Toronto, Laboratory Medicine and Pathobiology, Toronto, Canada; University of Toronto, Medicine, Toronto, Canada; University Health Network/Sinai Health Department of Microbiology, Toronto, Canada
| | - Julie L V Shaw
- Eastern Ontario Regional Laboratory Association, Canada; Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Canada
| | | | | | | |
Collapse
|
35
|
Kobayashi GS, Brito LA, Moreira DDP, Suzuki AM, Hsia GSP, Pimentel LF, de Paiva APB, Dias CR, Lourenço NCV, Oliveira BA, Manuli ER, Corral MA, Cavaçana N, Mitne-Neto M, Sales MM, Dell’ Aquila LP, Filho AR, Parrillo EF, Mendes-Corrêa MC, Sabino EC, Costa SF, Leal FE, Sgro GG, Farah CS, Zatz M, Passos-Bueno MR. A Novel Saliva RT-LAMP Workflow for Rapid Identification of COVID-19 Cases and Restraining Viral Spread. Diagnostics (Basel) 2021; 11:1400. [PMID: 34441334 PMCID: PMC8391450 DOI: 10.3390/diagnostics11081400] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/25/2021] [Accepted: 07/16/2021] [Indexed: 12/19/2022] Open
Abstract
Rapid diagnostics is pivotal to curb SARS-CoV-2 transmission, and saliva has emerged as a practical alternative to naso/oropharyngeal (NOP) specimens. We aimed to develop a direct RT-LAMP (reverse transcription loop-mediated isothermal amplification) workflow for viral detection in saliva, and to provide more information regarding its potential in curbing COVID-19 transmission. Clinical and contrived specimens were used to optimize formulations and sample processing protocols. Salivary viral load was determined in symptomatic patients to evaluate the clinical performance of the test and to characterize saliva based on age, gender and time from onset of symptoms. Our workflow achieved an overall sensitivity of 77.2% (n = 90), with 93.2% sensitivity, 97% specificity, and 0.895 Kappa for specimens containing >102 copies/μL (n = 77). Further analyses in saliva showed that viral load peaks in the first days of symptoms and decreases afterwards, and that viral load is ~10 times lower in females compared to males, and declines following symptom onset. NOP RT-PCR data did not yield relevant associations. This work suggests that saliva reflects the transmission dynamics better than NOP specimens, and reveals gender differences that may reflect higher transmission by males. This saliva RT-LAMP workflow can be applied to track viral spread and, to maximize detection, testing should be performed immediately after symptoms are presented, especially in females.
Collapse
Affiliation(s)
- Gerson Shigeru Kobayashi
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| | - Luciano Abreu Brito
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| | - Danielle de Paula Moreira
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| | - Angela May Suzuki
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| | - Gabriella Shih Ping Hsia
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| | - Lylyan Fragoso Pimentel
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| | - Ana Paula Barreto de Paiva
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| | - Carolina Regoli Dias
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| | - Naila Cristina Vilaça Lourenço
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| | - Beatriz Araujo Oliveira
- Instituto de Medicina Tropical, Universidade de São Paulo (USP), São Paulo 05403-000, Brazil; (B.A.O.); (E.R.M.); (M.C.M.-C.); (E.C.S.); (S.F.C.)
| | - Erika Regina Manuli
- Instituto de Medicina Tropical, Universidade de São Paulo (USP), São Paulo 05403-000, Brazil; (B.A.O.); (E.R.M.); (M.C.M.-C.); (E.C.S.); (S.F.C.)
| | - Marcelo Andreetta Corral
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| | - Natale Cavaçana
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| | - Miguel Mitne-Neto
- Grupo Fleury, Research and Development, São Paulo 04344-070, Brazil;
| | - Maria Mirtes Sales
- Instituto de Ensino e Pesquisa Prevent Senior, São Paulo 04547-100, Brazil; (M.M.S.); (L.P.D.A.); (A.R.F.); (E.F.P.)
| | - Luiz Phellipe Dell’ Aquila
- Instituto de Ensino e Pesquisa Prevent Senior, São Paulo 04547-100, Brazil; (M.M.S.); (L.P.D.A.); (A.R.F.); (E.F.P.)
| | - Alvaro Razuk Filho
- Instituto de Ensino e Pesquisa Prevent Senior, São Paulo 04547-100, Brazil; (M.M.S.); (L.P.D.A.); (A.R.F.); (E.F.P.)
| | - Eduardo Fagundes Parrillo
- Instituto de Ensino e Pesquisa Prevent Senior, São Paulo 04547-100, Brazil; (M.M.S.); (L.P.D.A.); (A.R.F.); (E.F.P.)
| | - Maria Cássia Mendes-Corrêa
- Instituto de Medicina Tropical, Universidade de São Paulo (USP), São Paulo 05403-000, Brazil; (B.A.O.); (E.R.M.); (M.C.M.-C.); (E.C.S.); (S.F.C.)
| | - Ester Cerdeira Sabino
- Instituto de Medicina Tropical, Universidade de São Paulo (USP), São Paulo 05403-000, Brazil; (B.A.O.); (E.R.M.); (M.C.M.-C.); (E.C.S.); (S.F.C.)
| | - Silvia Figueiredo Costa
- Instituto de Medicina Tropical, Universidade de São Paulo (USP), São Paulo 05403-000, Brazil; (B.A.O.); (E.R.M.); (M.C.M.-C.); (E.C.S.); (S.F.C.)
| | - Fabio Eudes Leal
- Faculdade de Medicina, Universidade Municipal de São Caetano do Sul (USCS), São Paulo 09521-160, Brazil;
| | - Germán Gustavo Sgro
- Instituto de Química, Universidade de São Paulo (USP), São Paulo 05508-000, Brazil; (G.G.S.); (C.S.F.)
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-903, Brazil
| | - Chuck Shaker Farah
- Instituto de Química, Universidade de São Paulo (USP), São Paulo 05508-000, Brazil; (G.G.S.); (C.S.F.)
| | - Mayana Zatz
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| | - Maria Rita Passos-Bueno
- Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco (HUG-CELL), Instituto de Biociências, Universidade de São Paulo (USP), São Paulo 05508-090, Brazil; (L.A.B.); (D.d.P.M.); (A.M.S.); (G.S.P.H.); (L.F.P.); (A.P.B.d.P.); (C.R.D.); (N.C.V.L.); (M.A.C.); (N.C.); (M.Z.)
| |
Collapse
|
36
|
Rose-Martel M, Tompkins E, Rutley R, Romero-Barrios P, Buenaventura E. Exposure Profile of Severe Acute Respiratory Syndrome Coronavirus 2 in Canadian Food Sources. J Food Prot 2021; 84:1295-1303. [PMID: 33770187 PMCID: PMC9805411 DOI: 10.4315/jfp-20-492] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/26/2021] [Indexed: 02/04/2023]
Abstract
ABSTRACT A new coronavirus strain known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread worldwide. This virus is the causative agent for coronavirus disease 2019 (COVID-19) and spreads primarily through human-to-human transmission via infected droplets and aerosols generated by infected persons. Although COVID-19 is a respiratory virus, the potential for transmission of SARS-CoV-2 via food is considered theoretically possible and remains a concern for Canadian consumers. We have conducted an exposure assessment of the likelihood of exposure of SARS-CoV-2 in Canadian food sources at the time of consumption. This article describes the exposure routes considered most relevant in the context of food contamination with SARS-CoV-2, including contaminated food of animal origin, other contaminated fresh foods, fomites, and SARS-CoV-2-contaminated feces. The likelihood of foodborne infection of SARS-CoV-2 via the human digestive tract also was considered. Our analysis indicates that there is no evidence that foodborne transmission of SARS-CoV-2 has occurred, and we consider the likelihood of contracting COVID-19 via food and food packaging in Canada as low to remote. Adherence to safe food practices and cleaning procedures would in any case prevent a potential foodborne infection with SARS-CoV-2. HIGHLIGHTS
Collapse
Affiliation(s)
- Megan Rose-Martel
- Bureau of Microbial Hazards, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Elizabeth Tompkins
- Bureau of Microbial Hazards, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Rebecca Rutley
- Bureau of Microbial Hazards, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Pablo Romero-Barrios
- Bureau of Microbial Hazards, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Enrico Buenaventura
- Bureau of Microbial Hazards, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| |
Collapse
|
37
|
Andriamandimby SF, Brook CE, Razanazatovo N, Rakotondramanga JM, Rasambainarivo F, Raharimanga V, Razanajatovo IM, Mangahasimbola R, Razafindratsimandresy R, Randrianarisoa S, Bernardson B, Rabarison JH, Randrianarisoa M, Nasolo FS, Rabetombosoa RM, Randremanana R, Héraud JM, Dussart P. Cross-sectional cycle threshold values reflect epidemic dynamics of COVID-19 in Madagascar. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.07.06.21259473. [PMID: 34268517 PMCID: PMC8282106 DOI: 10.1101/2021.07.06.21259473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
As the national reference laboratory for febrile illness in Madagascar, we processed samples from the first epidemic wave of COVID-19, between March and September 2020. We fit generalized additive models to cycle threshold (C t ) value data from our RT-qPCR platform, demonstrating a peak in high viral load, low-C t value infections temporally coincident with peak epidemic growth rates estimated in real time from publicly-reported incidence data and retrospectively from our own laboratory testing data across three administrative regions. We additionally demonstrate a statistically significant effect of duration of time since infection onset on C t value, suggesting that C t value can be used as a biomarker of the stage at which an individual is sampled in the course of an infection trajectory. As an extension, the population-level C t distribution at a given timepoint can be used to estimate population-level epidemiological dynamics. We illustrate this concept by adopting a recently-developed, nested modeling approach, embedding a within-host viral kinetics model within a population-level Susceptible-Exposed-Infectious-Recovered (SEIR) framework, to mechanistically estimate epidemic growth rates from cross-sectional C t distributions across three regions in Madagascar. We find that C t -derived epidemic growth estimates slightly precede those derived from incidence data across the first epidemic wave, suggesting delays in surveillance and case reporting. Our findings indicate that public reporting of C t values could offer an important resource for epidemiological inference in low surveillance settings, enabling forecasts of impending incidence peaks in regions with limited case reporting.
Collapse
|
38
|
Millar BC, Moore JE. Re-purposing of domestic steam disinfectors within the Hospital-at-Home setting: Reconciliation of steam disinfector thermal performance against SARS- CoV-2 (COVID-19), norovirus and other viruses' thermal susceptibilities. Infect Dis Health 2021; 26:156-159. [PMID: 33579632 PMCID: PMC7843083 DOI: 10.1016/j.idh.2021.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 11/19/2022]
Affiliation(s)
- Beverley C Millar
- Laboratory for Disinfection and Pathogen Elimination Studies, Northern Ireland Public Health Laboratory, Nightingale (Belfast City) Hospital, Lisburn Road, Belfast, Northern Ireland, BT9 7AD, UK; School of Medicine, Dentistry and Biomedical Science, The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK
| | - John E Moore
- Laboratory for Disinfection and Pathogen Elimination Studies, Northern Ireland Public Health Laboratory, Nightingale (Belfast City) Hospital, Lisburn Road, Belfast, Northern Ireland, BT9 7AD, UK; School of Medicine, Dentistry and Biomedical Science, The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK.
| |
Collapse
|
39
|
Lista MJ, Matos PM, Maguire TJA, Poulton K, Ortiz-Zapater E, Page R, Sertkaya H, Ortega-Prieto AM, O’Byrne AM, Bouton C, Dickenson RE, Ficarelli M, Jimenez-Guardeño JM, Howard M, Betancor G, Galao RP, Pickering S, Signell AW, Wilson H, Cliff P, Ik MTK, Patel A, MacMahon E, Cunningham E, Doores K, Agromayor M, Martin-Serrano J, Perucha E, Mischo HE, Shankar-Hari M, Batra R, Edgeworth J, Zuckerman M, Malim MH, Neil S, Martinez-Nunez RT. Resilient SARS-CoV-2 diagnostics workflows including viral heat inactivation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2020.04.22.20074351. [PMID: 33851184 PMCID: PMC8043481 DOI: 10.1101/2020.04.22.20074351] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There is a worldwide need for reagents to perform SARS-CoV-2 detection. Some laboratories have implemented kit-free protocols, but many others do not have the capacity to develop these and/or perform manual processing. We provide multiple workflows for SARS-CoV-2 nucleic acid detection in clinical samples by comparing several commercially available RNA extraction methods: QIAamp Viral RNA Mini Kit (QIAgen), RNAdvance Blood/Viral (Beckman) and Mag-Bind Viral DNA/RNA 96 Kit (Omega Bio-tek). We also compared One-step RT-qPCR reagents: TaqMan Fast Virus 1-Step Master Mix (FastVirus, ThermoFisher Scientific), qPCRBIO Probe 1-Step Go Lo-ROX (PCR Biosystems) and Luna ® Universal Probe One-Step RT-qPCR Kit (Luna, NEB). We used primer-probes that detect viral N (EUA CDC) and RdRP (PHE guidelines). All RNA extraction methods provided similar results. FastVirus and Luna proved most sensitive. N detection was more reliable than that of RdRP, particularly in samples with low viral titres. Importantly, we demonstrate that treatment of nasopharyngeal swabs with 70 degrees for 10 or 30 min, or 90 degrees for 10 or 30 min (both original variant and B 1.1.7) inactivates SARS-CoV-2 employing plaque assays, and that it has minimal impact on the sensitivity of the qPCR in clinical samples. These findings make SARS-CoV-2 testing portable to settings that do not have CL-3 facilities. In summary, we provide several testing pipelines that can be easily implemented in other laboratories and have made all our protocols and SOPs freely available at https://osf.io/uebvj/ .
Collapse
Affiliation(s)
- Maria Jose Lista
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
- All these authors contributed equally to the completion of this work
| | - Pedro M. Matos
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
- All these authors contributed equally to the completion of this work
| | - Thomas J. A. Maguire
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Inflammation Biology, School of Immunology and Microbial Sciences. Asthma UK Centre in Allergic Mechanisms of Asthma. Guy’s Campus, King’s College London SE1 9RT, UK
- All these authors contributed equally to the completion of this work
| | - Kate Poulton
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
- All these authors contributed equally to the completion of this work
| | - Elena Ortiz-Zapater
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Randall Centre for Cell & Molecular Biophysics. Guy’s Campus, King’s College London, SE1 1UL, UK
- Peter Gorer Department of Immunobiology. Guy’s Campus, King’s College London, SE1 9RT, UK
| | - Robert Page
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Inflammation Biology, School of Immunology and Microbial Sciences. Asthma UK Centre in Allergic Mechanisms of Asthma. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Helin Sertkaya
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Ana M. Ortega-Prieto
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Aoife M. O’Byrne
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Centre for Inflammation Biology and Cancer Immunology (CIBCI). Centre for Rheumatic Diseases (CRD – EULAR Centre of Excellence). Guy’s Campus, King’s College London SE1 1UL, UK
| | - Clement Bouton
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Ruth E Dickenson
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Mattia Ficarelli
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Jose M. Jimenez-Guardeño
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Mark Howard
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Peter Gorer Department of Immunobiology. Guy’s Campus, King’s College London, SE1 9RT, UK
| | - Gilberto Betancor
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Rui Pedro Galao
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Suzanne Pickering
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Adrian W Signell
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Harry Wilson
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | | | - Mark Tan Kia Ik
- Centre for Infectious Diseases Research, St Thomas’ Hospital (London, UK)
| | - Amita Patel
- Centre for Infectious Diseases Research, St Thomas’ Hospital (London, UK)
| | - Eithne MacMahon
- Centre for Infectious Diseases Research, St Thomas’ Hospital (London, UK)
| | - Emma Cunningham
- Centre for Infectious Diseases Research, St Thomas’ Hospital (London, UK)
| | - Katie Doores
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Monica Agromayor
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Juan Martin-Serrano
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Esperanza Perucha
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Centre for Inflammation Biology and Cancer Immunology (CIBCI). Centre for Rheumatic Diseases (CRD – EULAR Centre of Excellence). Guy’s Campus, King’s College London SE1 1UL, UK
| | - Hannah E. Mischo
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Manu Shankar-Hari
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Rahul Batra
- Centre for Infectious Diseases Research, St Thomas’ Hospital (London, UK)
| | - Jonathan Edgeworth
- Centre for Infectious Diseases Research, St Thomas’ Hospital (London, UK)
| | - Mark Zuckerman
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Virology. King’s College Hospital (London, UK)
| | - Michael H. Malim
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Stuart Neil
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| | - Rocio Teresa Martinez-Nunez
- King’s College London Diagnostics Team at Guy’s Campus (London, UK)
- Dept. Infectious Diseases, School of Immunology and Microbial Sciences. Guy’s Campus, King’s College London SE1 9RT, UK
| |
Collapse
|
40
|
Kwok CS, Dashti M, Tafuro J, Nasiri M, Muntean EA, Wong N, Kemp T, Hills G, Mallen CD. Methods to disinfect and decontaminate SARS-CoV-2: a systematic review of in vitro studies. Ther Adv Infect Dis 2021; 8:2049936121998548. [PMID: 33796289 PMCID: PMC7970236 DOI: 10.1177/2049936121998548] [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: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Cleaning is a major control component for outbreaks of infection. However, for the SARS-CoV-2 pandemic, there is limited specific guidance regarding the proper disinfection methods that should be used. METHODS We conducted a systematic review of the literature on cleaning, disinfection or decontamination methods in the prevention of SARS-CoV-2. RESULTS A total of 27 studies were included, reporting a variety of methods with which the effectiveness of interventions were assessed. Virus was inoculated onto different types of material including masks, nasopharyngeal swabs, serum, laboratory plates and simulated saliva, tears or nasal fluid and then interventions were applied in an attempt to eliminate the virus including chemical, ultraviolet (UV) light irradiation, and heat and humidity. At body temperature (37°C) there is evidence that the virus will not be detectable after 2 days but this can be reduced to non-detection at 30 min at 56°C, 15 min at 65°C and 2 min at 98°C. Different experimental methods testing UV light have shown that it can inactivate the virus. Light of 254-365 nm has been used, including simulated sunlight. Many chemical agents including bleach, hand sanitiser, hand wash, soap, ethanol, isopropanol, guandinium thiocynate/t-octylphenoxypolyethoxyethanol, formaldehyde, povidone-iodine, 0.05% chlorhexidine, 0.1% benzalkonium chloride, acidic electrolysed water, Clyraguard copper iodine complex and hydrogen peroxide vapour have been shown to disinfect SARS-CoV-2. CONCLUSIONS Heating, UV light irradiation and chemicals can be used to inactivate SARS-CoV-2 but there is insufficient evidence to support one measure over others in clinical practice.
Collapse
Affiliation(s)
- Chun Shing Kwok
- Department of Cardiology, Royal Stoke University
Hospital, Stoke-on-Trent, UK
- School of Medicine, Keele University,
Stoke-on-Trent, UK
| | | | - Jacopo Tafuro
- Department of Cardiology, Royal Stoke University
Hospital, Stoke-on-Trent, UK
| | - Mojtaba Nasiri
- School of Life Sciences, University of Sussex,
Brighton, UK
| | | | - Nicholas Wong
- Department of Infectious Disease, Leicester
Royal Infirmary, Leicester, UK
| | - Timothy Kemp
- Department of Infectious Disease, Royal Stoke
University Hospital, Stoke-on-Trent, UK
| | - George Hills
- Department of Infectious Disease, Leicester
Royal Infirmary, Leicester, UK
| | | |
Collapse
|