1
|
Kumar S, Bhowmik B. EffiCOVID-net: A highly efficient convolutional neural network for COVID-19 diagnosis using chest X-ray imaging. Methods 2025; 240:81-100. [PMID: 40252941 DOI: 10.1016/j.ymeth.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025] Open
Abstract
The global COVID-19 pandemic has drastically affected daily life, emphasizing the urgent need for early and accurate detection to provide adequate medical treatment, especially with limited antiviral options. Chest X-ray imaging has proven crucial for distinguishing COVID-19 from other respiratory conditions, providing an essential diagnostic tool. Deep learning (DL)-based models have proven highly effective in image diagnostics in recent years. Many of these models are computationally intensive and prone to overfitting, especially when trained on limited datasets. Additionally, conventional models often fail to capture multi-scale features, reducing diagnostic accuracy. This paper proposed a highly efficient convolutional neural network (CNN) called EffiCOVID-Net, incorporating diverse feature learning units. The proposed model consists of a bunch of EffiCOVID blocks that incorporate several layers of convolution containing (3×3) filters and recurrent connections to extract complex features while preserving spatial integrity. The performance of EffiCOVID-Net is rigorously evaluated using standard performance metrics on two publicly available COVID-19 chest X-ray datasets. Experimental results demonstrate that EffiCOVID-Net outperforms existing models, achieving 98.68% accuracy on the COVID-19 radiography dataset (D1), 98.55% on the curated chest X-ray dataset (D2), and 98.87% on the mixed dataset (DMix) in multi-class classification (COVID-19 vs. Normal vs. Pneumonia). For binary classification (COVID-19 vs. Normal), the model attains 99.06%, 99.78%, and 99.07% accuracy, respectively. Integrating Grad-CAM-based visualizations further enhances interpretability by highlighting critical regions influencing model predictions. EffiCOVID-Net's lightweight architecture ensures low computational overhead, making it suitable for deployment in resource-constrained clinical settings. A comparative analysis with existing methods highlights its superior accuracy, efficiency, and robustness performance. However, while the model enhances diagnostic workflows, it is best utilized as an assistive tool rather than a standalone diagnostic method.
Collapse
Affiliation(s)
- Sunil Kumar
- Maharshi Patanjali CPS Lab, BRICS Laboratory, Department of Computer Science and Engineering, National Institute of Technology Karnataka, Mangalore 575025, Karnataka, India.
| | - Biswajit Bhowmik
- Maharshi Patanjali CPS Lab, BRICS Laboratory, Department of Computer Science and Engineering, National Institute of Technology Karnataka, Mangalore 575025, Karnataka, India
| |
Collapse
|
2
|
Dai C, Li Q, Wang L, Zhang J, Yang S, Zhang X. Long Noncoding LINC00115 Facilitates Cell Growth and Inhibits Apoptosis by Regulating the miR-4701-5p/P4HB Axis in Bladder Cancer. TOHOKU J EXP MED 2025; 265:69-81. [PMID: 39111879 DOI: 10.1620/tjem.2024.j075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Bladder cancer (BCa) is a prevalent urogenital malignancy, imposing a significant burden on health-care systems worldwide. Long noncoding RNAs (lncRNAs) are important regulators of carcinogenesis and affect BCa progression. In this study, the influence of lncRNA LINC00115 on malignant behavior of BCa cells were explored. Bioinformatics method was used for prediction of gene expression and downstream molecules of LIN00115. LINC00115 expression level in BCa cells was measured using RT-qPCR. After LINC00115 depletion, the proportion of viable, proliferative, and apoptotic BCa cells were calculated by methyl thiazolyl tetrazolium (MTT) assays, colony formation assays, and TUNEL staining, respectively. FISH was performed to verify the cellular distribution of LINC00115. The interaction between LINC00115 and miR-4701-5p and the binding between miR-4701-5p and P4HB were confirmed using RNA pulldown, RNA immunoprecipitation (RIP), and luciferase reporter assays. Experimental results showed that LINC00115 was highly expressed in BCa cells. The silencing of LINC00115 restrained BCa cell proliferation and stimulated apoptosis. LINC00115 could directly bind to miR-4701-5p and thus initiate P4HB upregulation in BCa cells. P4HB 3'untranslated region could be targeted by miR-4701-5p. Additionally, Amplification of P4HB expression offset the effects of LINC00115 knockdown on BCa cell proliferative and apoptotic behaviors. In conclusion, LINC00115 facilitates BCa cell growth and inhibits apoptosis via interaction with miR-4701-5p and upregulation of P4HB.
Collapse
Affiliation(s)
- Changyuan Dai
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| | - Qingwen Li
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| | - Lili Wang
- Department of Emergency medicine, The First Affiliated Hospital of Bengbu Medical College
| | - Jiajun Zhang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| | - Shuai Yang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| | - Xiaole Zhang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| |
Collapse
|
3
|
Odiwuor N, Li J, He P, Wang N, Murtaza A, Jiang M, Yu J, Wei H. Facilitating self-testing with a fast, accurate, and simplified shelf-stable colorimetric LAMP system for Mpox and SARS-CoV-2 detection. Talanta 2025; 283:127119. [PMID: 39509899 DOI: 10.1016/j.talanta.2024.127119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/23/2024] [Accepted: 10/27/2024] [Indexed: 11/15/2024]
Abstract
The rapid and accurate detection of viral infections is essential for effective disease management and prevention. Quantitative polymerase chain reaction (qPCR) remains the gold standard for viral detection due to its high sensitivity and specificity. However, its limitations-including the need for specialized equipment, trained personnel, and longer processing times-make it impractical for at-home or rapid testing. Although numerous point-of-care assays based on isothermal nucleic acid amplification have been developed, they often lack the simplicity and adaptability required for self-testing in non-laboratory settings such as at home. To address this, we developed and validated the SCOLAR (Shelf-stable Colorimetric LAMP system for Rapid self-testing of viruses) system, a simplified, portable, and accurate diagnostic tool designed for self-testing of Mpox and SARS-CoV-2 infections. The SCOLAR system employs novel lyophilized colorimetric loop-mediated isothermal amplification (LAMP) beads, a customized sample lysis buffer, and smartphone-assisted RGB color analysis for interpreting results. Validation was conducted using 24 mock Mpox skin swabs, 32 wastewater samples, and 104 clinical SARS-CoV-2 nasopharyngeal swabs, with comparisons to an in-house qPCR assay. The SCOLAR system demonstrated an analytical sensitivity of below 10 copies/μL for all targets within 15 min. Diagnostic performance for mock Mpox samples exhibited 93.8 % sensitivity and 100 % specificity, while wastewater samples achieved 100 % sensitivity and specificity. SARS-CoV-2 swabs had 96 % sensitivity and 100 % specificity. The system also proved effective for self-testing by untrained individuals. SCOLAR offers a reliable, easy-to-use platform for rapid self-testing, with potential for broader applications in public health strategies to enhance pandemic preparedness and response.
Collapse
Affiliation(s)
- Nelson Odiwuor
- WHP Innovation Lab, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430207, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Sino-Africa Joint Research Centre, Nairobi, 62000 - 00200, Kenya
| | - Junhua Li
- WHP Innovation Lab, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
| | - Ping He
- WHP Innovation Lab, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430207, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Nuo Wang
- WHP Innovation Lab, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
| | - Ali Murtaza
- WHP Innovation Lab, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430207, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mengwei Jiang
- WHP Innovation Lab, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
| | - Junping Yu
- WHP Innovation Lab, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
| | - Hongping Wei
- WHP Innovation Lab, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430207, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Sino-Africa Joint Research Centre, Nairobi, 62000 - 00200, Kenya.
| |
Collapse
|
4
|
Chwa JS, Shin Y, Lee Y, Fabrizio T, Congrave-Wilson Z, Cheng WA, Jumarang J, Kim M, Webby R, Bender JM, Pannaraj PS. SARS-CoV-2 Variants May Affect Saliva RT-PCR Assay Sensitivity. J Appl Lab Med 2024; 9:927-937. [PMID: 39246012 PMCID: PMC12001863 DOI: 10.1093/jalm/jfae095] [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: 02/07/2024] [Accepted: 07/09/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants demonstrate predilection for different regions of the respiratory tract. While saliva-based reverse transcription-polymerase chain reaction (RT-PCR) testing is a convenient, cost-effective alternative to nasopharyngeal swabs (NPS), few studies to date have investigated whether saliva sensitivity differs across variants of concern. METHODS SARS-CoV-2 RT-PCR was performed on paired NPS and saliva specimens collected from individuals with acute coronavirus disease 2019 (COVID-19) symptoms or exposure to a COVID-19 household contact. Viral genome sequencing of NPS specimens and Los Angeles County surveillance data were used to determine the variant of infection. Saliva sensitivity was calculated using NPS-positive RT-PCR as the reference standard. Factors contributing to the likelihood of saliva SARS-CoV-2 RT-PCR positivity were evaluated with univariate and multivariable analyses. RESULTS Between June 2020 and December 2022, 548 saliva samples paired with SARS-CoV-2 positive NPS samples were tested by RT-PCR. Overall, saliva sensitivity for SARS-CoV-2 detection was 61.7% (95% CI, 57.6%-65.7%). Sensitivity was highest with Delta infection (79.6%) compared to pre-Delta (58.5%) and Omicron (61.5%) (P = 0.003 and 0.01, respectively). Saliva sensitivity was higher in symptomatic individuals across all variants compared to asymptomatic cases [pre-Delta 80.6% vs 48.3% (P < 0.001), Delta 100% vs 72.5% (P = 0.03), Omicron 78.7% vs 51.2% (P < 0.001)]. Infection with Delta, symptoms, and high NPS viral load were independently associated with 2.99-, 3.45-, and 4.0-fold higher odds of SARS-CoV-2 detection by saliva-based RT-PCR (P = 0.004, <0.001, and <0.001), respectively. CONCLUSIONS As new variants emerge, evaluating saliva-based testing approaches may be crucial to ensure effective virus detection.
Collapse
Affiliation(s)
- Jason S. Chwa
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Yunho Shin
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Yesun Lee
- Division of Infectious Diseases, Department of Pediatrics, University of California, San Diego, California, USA
| | - Thomas Fabrizio
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Zion Congrave-Wilson
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Wesley A. Cheng
- Division of Infectious Diseases, Department of Pediatrics, University of California, San Diego, California, USA
| | - Jaycee Jumarang
- Division of Infectious Diseases, Department of Pediatrics, University of California, San Diego, California, USA
| | - Minjun Kim
- Division of Infectious Diseases, Department of Pediatrics, University of California, San Diego, California, USA
| | - Richard Webby
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jeffrey M. Bender
- Department of Pediatrics, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Pia S. Pannaraj
- Division of Infectious Diseases, Department of Pediatrics, University of California, San Diego, California, USA
| |
Collapse
|
5
|
Yoshikawa AAG, Cardoso SF, Eslabão LB, Pinheiro IC, Valverde P, Caminha G, Romero OB, Medeiros L, Rona LDP, Pitaluga AN. Point-of-care testing for COVID-19: a simple two-step molecular diagnostic development and validation during the SARS-CoV-2 pandemic. Mem Inst Oswaldo Cruz 2024; 119:e230236. [PMID: 39383402 PMCID: PMC11452069 DOI: 10.1590/0074-02760230236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 09/04/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND During the coronavirus disease 19 (COVID-19) pandemic, diagnostic testing of the general population proved challenging due to limitations of the gold-standard diagnostic procedure using reverse transcription real-time polymerase chain reaction (RT-qPCR) for large-scale testing on the centralised model, especially in low-resource areas. OBJECTIVES To address this, a point-of-care (PoC) diagnostic protocol for COVID-19 was developed, providing fast, reliable, and affordable testing, particularly for low-mid develop areas. METHODS The PoC diagnostic process combines a simple paper-based RNA extraction method housed within a 3D-printed plastic device with a colorimetric reverse transcription loop-mediated isothermal amplification (RT-LAMP) assay. Nasopharyngeal/oropharyngeal swabs (NOS) and saliva samples were tested between 2020 and 2021, with the assistance of Santa Catarina's State Health Secretary, Brazil. FINDINGS The developed diagnostic protocol showed a limit of detection of 9,900 copies and an overall diagnostic specificity of 98% for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from 1,348 clinical analysed samples. The diagnostic sensitivity was 95% for NOS samples, 85% for early morning saliva, and 69% for indiscriminate saliva. MAIN CONCLUSIONS In conclusion, the developed device successfully extracted SARS-CoV-2 viral RNA from swabs and saliva clinical samples. When combined with colorimetric RT-LAMP, it provides results within 45 min using minimal resources, thus delivering a diagnostic kit protocol that is applicable in large-scale sampling.
Collapse
Affiliation(s)
- Andre Akira Gonzaga Yoshikawa
- Universidade Federal de Santa Catarina, Departamento de Biologia Celular, Embriologia e Genética, Florianópolis, SC, Brasil
| | - Sabrina Fernandes Cardoso
- Universidade Federal de Santa Catarina, Departamento de Biologia Celular, Embriologia e Genética, Florianópolis, SC, Brasil
- Secretaria de Saúde do Estado de Santa Catarina, Diretoria de Vigilância Epidemiológica, Florianópolis, SC, Brasil
| | - Lívia Budziarek Eslabão
- Universidade Federal de Santa Catarina, Departamento de Microbiologia, Imunologia e Parasitologia, Florianópolis, SC, Brasil
| | - Iara Carolini Pinheiro
- Universidade Federal de Santa Catarina, Departamento de Biologia Celular, Embriologia e Genética, Florianópolis, SC, Brasil
| | | | - Gisele Caminha
- Laboratório Central de Saúde Pública de Santa Catarina, Florianópolis, SC, Brasil
| | - Oscar Bruna Romero
- Universidade Federal de Santa Catarina, Departamento de Microbiologia, Imunologia e Parasitologia, Florianópolis, SC, Brasil
| | - Leandro Medeiros
- Instituto Federal de Educação, Ciência e Tecnologia de Santa Catarina, Florianópolis, SC, Brasil
| | - Luísa Damazio Pitaluga Rona
- Universidade Federal de Santa Catarina, Departamento de Biologia Celular, Embriologia e Genética, Florianópolis, SC, Brasil
- Conselho Nacional de Desenvolvimento Científico e Tecnológico, Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brasil
| | - André Nóbrega Pitaluga
- Conselho Nacional de Desenvolvimento Científico e Tecnológico, Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brasil
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
6
|
Hirabayashi E, Mercado G, Hull B, Soin S, Koshy-Chenthittayil S, Raman S, Huang T, Keerthisinghe C, Feliciano S, Dongo A, Kal J, Azizan A, Duus K, Else T, DeArmond M, Stone AE. Comparison of diagnostic accuracy of rapid antigen tests for COVID-19 compared to the viral genetic test in adults: a systematic review and meta-analysis. JBI Evid Synth 2024; 22:1939-2002. [PMID: 39188132 PMCID: PMC11462910 DOI: 10.11124/jbies-23-00291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
OBJECTIVE The objective of this review was to determine the diagnostic accuracy of the currently available and upcoming point-of-care rapid antigen tests (RATs) used in primary care settings relative to the viral genetic real-time reverse transcriptase polymerase chain reaction (RT-PCR) test as a reference for diagnosing COVID-19/SARS-CoV-2 in adults. INTRODUCTION Accurate COVID-19 point-of-care diagnostic tests are required for real-time identification of SARS-CoV-2 infection in individuals. Real-time RT-PCR is the accepted gold standard for diagnostic testing, requiring technical expertise and expensive equipment that are unavailable in most primary care locations. RATs are immunoassays that detect the presence of a specific viral protein, which implies a current infection with SARS-CoV-2. RATs are qualitative or semi-quantitative diagnostics that lack thresholds that provide a result within a short time frame, typically within the hour following sample collection. In this systematic review, we synthesized the current evidence regarding the accuracy of RATs for detecting SARS-CoV-2 compared with RT-PCR. INCLUSION CRITERIA Studies that included nonpregnant adults (18 years or older) with suspected SARS-CoV-2 infection, regardless of symptomology or disease severity, were included. The index test was any available SARS-CoV-2 point-of-care RAT. The reference test was any commercially distributed RT-PCR-based test that detects the RNA genome of SARS-CoV-2 and has been validated by an independent third party. Custom or in-house RT-PCR tests were also considered, with appropriate validation documentation. The diagnosis of interest was COVID-19 disease and SARS-CoV-2 infection. This review considered cross-sectional and cohort studies that examined the diagnostic accuracy of COVID-19/SARS-CoV-2 infection where the participants had both index and reference tests performed. METHODS The keywords and index terms contained in relevant articles were used to develop a full search strategy for PubMed and adapted for Embase, Scopus, Qinsight, and the WHO COVID-19 databases. Studies published from November 2019 to July 12, 2022, were included, as SARS-CoV-2 emerged in late 2019 and is the cause of a continuing pandemic. Studies that met the inclusion criteria were critically appraised using QUADAS-2. Using a customized tool, data were extracted from included studies and were verified prior to analysis. The pooled sensitivity, specificity, positive predictive, and negative predictive values were calculated and presented with 95% CIs. When heterogeneity was observed, outlier analysis was conducted, and the results were generated by removing outliers. RESULTS Meta-analysis was performed on 91 studies of 581 full-text articles retrieved that provided true-positive, true-negative, false-positive, and false-negative values. RATs can identify individuals who have COVID-19 with high reliability (positive predictive value 97.7%; negative predictive value 95.2%) when considering overall performance. However, the lower level of sensitivity (67.1%) suggests that negative test results likely need to be retested through an additional method. CONCLUSIONS Most reported RAT brands had only a few studies comparing their performance with RT-PCR. Overall, a positive RAT result is an excellent predictor of a positive diagnosis of COVID-19. We recommend that Roche's SARS-CoV-2 Rapid Antigen Test and Abbott's BinaxNOW tests be used in primary care settings, with the understanding that negative results need to be confirmed through RT-PCR. We recommend adherence to the STARD guidelines when reporting on diagnostic data. REVIEW REGISTRATION PROSPERO CRD42020224250.
Collapse
Affiliation(s)
- Ellyn Hirabayashi
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Guadalupe Mercado
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Brandi Hull
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Sabrina Soin
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Sherli Koshy-Chenthittayil
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Sarina Raman
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Timothy Huang
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Chathushya Keerthisinghe
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Shelby Feliciano
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Andrew Dongo
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - James Kal
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Azliyati Azizan
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Karen Duus
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Terry Else
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| | - Megan DeArmond
- Touro University Nevada, Jay Sexter Library, Henderson, NV, USA
- Touro University Nevada: JBI Affiliated Group, Henderson, NV, USA
| | - Amy E.L. Stone
- Touro University Nevada, College of Osteopathic Medicine, Department of Basic Sciences, Henderson, NV, USA
| |
Collapse
|
7
|
Sabateen A, Sadaqa D, Nino T, Zaghal G, Qumsieh G, Fakhori R, Rjoub H, Taha T, Zghari R, Abu Hanieh S, Al-Basha D, Qabaja M, Alsaid H, Hindiyeh MY. Evaluation of COVID-19 rapid antigen test against polymerase chain reaction test in immunocompromised patients. PLoS One 2024; 19:e0306396. [PMID: 39093858 PMCID: PMC11296623 DOI: 10.1371/journal.pone.0306396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/14/2024] [Indexed: 08/04/2024] Open
Abstract
On the 11th of March 2020, the world faced a new global pandemic, COVID-19 which is a disease caused by the novel coronavirus, it had multiple devastating outcomes on multiple sectors along with significant rates of mortality. These challenges encouraged the development of multiple testing methods, as well as anti-viral medications such as Molnupiravir, as well as evaluating the efficacy of available medications against it, like; Azithromycin, Ritonavir and Hydroxychloroquine. Vaccination against COVID-19 forged into a significant challenge, few months ensuing the first case of SARS-CoV-2, which was diagnosed in December 2019, in Wuhan-China, thus, multiple vaccines were approved for use around the world to combat this pandemic. Our study includes a sample of 556 oncology patients at Augusta Victoria Hospital in Jerusalem, all patients were tested using Panbio rapid antigen test and Allplex PCR Assay. The main objective was to study the sensitivity and specificity of Rapid antigen test, which contributes to a faster isolation call and management of infected patients, thus decreasing the risk on spread to other patients and health care. Patients were categorized based on two factors: Ct range and age group and studying their possible effect on false-negative results. Patients with Ct value less than 20, had the highest detection rate which is consistent with other studies in the literature. The sensitivity and specificity of Panbio Rapid Antigen testing were of 69.9% and 100%, respectively. A correlation between age group and false negative results could not be made, but a correlation between Ct value and false negative result was noticed, Ct value was directly related to false negative results. P-value of 0.007 indicated that results were statistically significant where PCR test is considered more sensitive compared to rapid antigen test.
Collapse
Affiliation(s)
- Ali Sabateen
- Infectious Diseases Unit, Augusta Victoria Hospital, East Jerusalem, Palestine
| | - Dana Sadaqa
- Infectious Diseases Unit, Augusta Victoria Hospital, East Jerusalem, Palestine
| | - Taleen Nino
- Molecular Laboratory, Augusta Victoria Hospital, East Jerusalem, Palestine
| | - Ghayd Zaghal
- Molecular Laboratory, Augusta Victoria Hospital, East Jerusalem, Palestine
| | - George Qumsieh
- Molecular Laboratory, Augusta Victoria Hospital, East Jerusalem, Palestine
| | - Reena Fakhori
- Molecular Laboratory, Augusta Victoria Hospital, East Jerusalem, Palestine
| | - Hammam Rjoub
- Infectious Diseases Unit, Augusta Victoria Hospital, East Jerusalem, Palestine
| | - Tahreer Taha
- Infectious Diseases Unit, Augusta Victoria Hospital, East Jerusalem, Palestine
| | - Rami Zghari
- Infectious Diseases Unit, Augusta Victoria Hospital, East Jerusalem, Palestine
| | - Sari Abu Hanieh
- Infectious Diseases Unit, Augusta Victoria Hospital, East Jerusalem, Palestine
| | - Duaa Al-Basha
- Infectious Diseases Unit, Augusta Victoria Hospital, East Jerusalem, Palestine
- Pharmacy Department, Modern University College (MUC), Ramallah, Palestine
| | - Marwan Qabaja
- Pathology Laboratory, Augusta Victoria Hospital, East Jerusalem, Palestine
| | - Hamza Alsaid
- Internal Medicine Department, Hadassah University Hospital, Jerusalem, Israel
| | - Musa Y. Hindiyeh
- Molecular Laboratory, Augusta Victoria Hospital, East Jerusalem, Palestine
| |
Collapse
|
8
|
Turbawaty DK, Sudjadi A, Lismayanti L, Rostini T, Logito V. The Performance of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) Virus Using the Colorimetric Reverse-Transcription Loop Mediated Isothermal Amplification (RT-LAMP) Method in Saliva Specimens of Suspected COVID-19 Patients. Int J Gen Med 2024; 17:3329-3335. [PMID: 39081671 PMCID: PMC11288315 DOI: 10.2147/ijgm.s461613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/09/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction Corona Virus Disease-19 (COVID-19) is a disease caused by Severe-Acute-Respiratory-Syndrome-Coronavirus-2 (SARS-CoV-2). The most reliable and widely accepted method for diagnosing this infection, despite facing various challenges, is the Reverse Transcription Polymerase Chain Reaction (RT-PCR) method, which utilizes nasopharyngeal swab sample. Reverse-transcription loop mediated isothermal amplification (RT-LAMP) is a simpler nucleic acid amplification method compared to the RT-PCR method. This method has several advantages, including: of amplification at constant temperature, faster results, and potentially greater examination capacity. Purpose This study aimed to compare the validity of the RT-LAMP method using saliva specimens with that of the RT-PCR method using nasopharyngeal smears. Methods This was an analytical observational study with a cross-sectional design. The participants were inpatients in the COVID-19 special isolation building of Hasan Sadikin General Hospital, Indonesia with a probable (clinical symptoms of covid, but not confirm NAAT examination) or confirmed diagnosis of COVID-19 from September 2021 to February 2022. The inclusion criteria are COVID-19 patients with symptoms, adult subjects, and composite mentions. Patients who were unable to secrete saliva were also excluded. Results In total, 118 specimens were collected. The validity test results of the saliva specimens using the RT-LAMP method showed sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV), of 65.5%, 100%, 100%, and 75%, respectively. The results increased in subjects treated between 3 and 7 days after symptom onset ie 73.2%, 100%, 100%, and 82.3%, respectively. Conclusion The very strong specificity accompanied by good sensitivity and NPV in the group of subjects treated 3-7 days after the onset of symptoms indicates that the RT-LAMP method using saliva specimens can be an efficient and reliable alternative tool in detecting the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Dewi Kartika Turbawaty
- Department of Clinical Pathology, Faculty of Medicine Padjadjaran University/ Dr. Hasan Sadikin General Hospital, Bandung, West Java, Indonesia
| | - Andy Sudjadi
- Department of Clinical Pathology, Faculty of Medicine Padjadjaran University/ Dr. Hasan Sadikin General Hospital, Bandung, West Java, Indonesia
| | - Leni Lismayanti
- Department of Clinical Pathology, Faculty of Medicine Padjadjaran University/ Dr. Hasan Sadikin General Hospital, Bandung, West Java, Indonesia
| | - Tiene Rostini
- Department of Clinical Pathology, Faculty of Medicine Padjadjaran University/ Dr. Hasan Sadikin General Hospital, Bandung, West Java, Indonesia
| | - Verina Logito
- Department of Clinical Pathology, Faculty of Medicine Padjadjaran University/ Dr. Hasan Sadikin General Hospital, Bandung, West Java, Indonesia
| |
Collapse
|
9
|
Samsunder N, Sivro A, Hassan-Moosa R, Lewis L, Kara Z, Baxter C, Karim QA, Karim SA, Kharsany ABM, Naidoo K, Ngcapu S. Evaluating diagnostic accuracy of an RT-PCR test for the detection of SARS-CoV-2 in saliva. Diagn Progn Res 2024; 8:9. [PMID: 39044271 PMCID: PMC11267770 DOI: 10.1186/s41512-024-00176-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 07/01/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Saliva has been proposed as a potential more convenient, cost-effective, and easier sample for diagnosing SARS-CoV-2 infections, but there is limited knowledge of the impact of saliva volumes and stages of infection on its sensitivity and specificity. METHODS In this study, we assessed the performance of SARS-CoV-2 testing in 171 saliva samples from 52 mostly mildly symptomatic patients (aged 18 to 70 years) with a positive reference standard result at screening. The samples were collected at different volumes (50, 100, 300, and 500 µl of saliva) and at different stages of the disease (at enrollment, day 7, 14, and 28 post SARS-CoV-2 diagnosis). Imperfect nasopharyngeal (NP) swab nucleic acid amplification testing was used as a reference. We used a logistic regression with generalized estimating equations to estimate sensitivity, specificity, PPV, and NPV, accounting for the correlation between repeated observations. RESULTS The sensitivity and specificity values were consistent across saliva volumes. The sensitivity of saliva samples ranged from 70.2% (95% CI, 49.3-85.0%) for 100 μl to 81.0% (95% CI, 51.9-94.4%) for 300 μl of saliva collected. The specificity values ranged between 75.8% (95% CI, 55.0-88.9%) for 50 μl and 78.8% (95% CI, 63.2-88.9%) for 100 μl saliva compared to NP swab samples. The overall percentage of positive results in NP swabs and saliva specimens remained comparable throughout the study visits. We observed no significant difference in cycle number values between saliva and NP swab specimens, irrespective of saliva volume tested. CONCLUSIONS The saliva collection offers a promising approach for population-based testing.
Collapse
Affiliation(s)
- Natasha Samsunder
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
| | - Aida Sivro
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
- JC Wilt Infectious Disease Research Centre, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Razia Hassan-Moosa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
- SAMRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Lara Lewis
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
| | - Zahra Kara
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
| | - Cheryl Baxter
- Centre for Epidemic Response and Innovation (CERI), Stellenbosch University, Stellenbosch, South Africa
| | - Quarraisha Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Salim Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Ayesha B M Kharsany
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa
- SAMRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Sinaye Ngcapu
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), 719 Umbilo Road, Durban, 4001, South Africa.
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
10
|
Takeuchi JS, Fukano K, Kito Y, Yamamoto K, Kimura M. Evaluation of the SARS-CoV-2 RNA detection reagent LAMPdirect Genelyzer KIT using nasopharyngeal swab and saliva samples. Diagn Microbiol Infect Dis 2024; 109:116297. [PMID: 38604076 DOI: 10.1016/j.diagmicrobio.2024.116297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
The LAMPdirect Genelyzer KIT allows for the detection of SARS-CoV-2 RNA in saliva samples with a loop-mediated isothermal amplification (LAMP) method and generates results within 20 min. It has been approved by the Pharmaceuticals and Medical Devices Agency in Japan. In this study, the performance of the LAMPdirect Genelyzer KIT was compared with that of the RT-qPCR reference method using 50 nasopharyngeal swabs and 100 saliva samples. In addition, we evaluated the applicability of an alternative reverse transcriptase and the effects of an inactivation buffer. The total agreement rates were 80.0 % and 82.0 % for nasopharyngeal and saliva samples, respectively. When considering samples at the detection limit (50 copies/reaction) that increases the chance of transmission between humans, the total agreement rates were 100% and 94.1% for nasopharyngeal and saliva samples, respectively. The LAMP method is simple, fast, and inexpensive, making it useful for small medical institutions or rural areas.
Collapse
Affiliation(s)
- Junko S Takeuchi
- Department of Academic-Industrial Partnerships Promotion, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo 162-8655, Japan.
| | - Kento Fukano
- Department of Academic-Industrial Partnerships Promotion, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo 162-8655, Japan.
| | - Yumiko Kito
- Department of Academic-Industrial Partnerships Promotion, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Kei Yamamoto
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo 162-8655, Japan.
| | - Moto Kimura
- Department of Academic-Industrial Partnerships Promotion, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo 162-8655, Japan.
| |
Collapse
|
11
|
Herrmann L, Breuer J, Duc TN, Thomé N, Ghazaani F, Kamhieh-Milz S, Kamhieh-Milz J, Pfützner A. Comparison of the diagnostic accuracy of the Pluslife Mini Dock RHAM technology with Abbott ID Now and Cepheid GenXpert: A retrospective evaluation study. Sci Rep 2024; 14:13978. [PMID: 38886535 PMCID: PMC11183097 DOI: 10.1038/s41598-024-64406-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
Rapid and sensitive detection of pathogens is critical in interrupting the transmission chain of infectious diseases. Currently, real-time (RT-)PCR represents the gold standard for the detection of SARS-CoV-2. RNase HII-assisted amplification (RHAM) is a promising technology, enabling reliable point-of-care (PoC) testing; however, its diagnostic accuracy has not yet been investigated. The present study compared the Pluslife Mini Dock (RHAM technology), with Abbott ID Now and Cepheid GeneXpert IV. The positive percent agreement (PPA) and negative percent agreement (NPA) were determined in 100 SARS-CoV-2 positive and 210 SARS-CoV-2 negative samples. Further, the reliability of the Pluslife Mini Dock was investigated in different SARS-CoV-2 variants (Delta and Omicron subvariants). The PPA was 99.00% for Pluslife, 100.00% for Abbott ID Now, and 99.00% for Cepheid GeneXpert, with an NPA of 100.00%, 98.90%, and 93.72%, respectively. Abbott ID Now demonstrated the highest rate of invalid results. All SARS-CoV-2 analysed variants were detected by the Pluslife device. Altogether, the Pluslife Mini Dock demonstrated a PPA of 99.16% (235/237) for CT < 36 and an NPA of 100.00% (313/313), respectively. In conclusion, the Pluslife Mini Dock demonstrated better analytical performance than Abbott ID Now and Cepheid GeneXpert IV, representing a highly accurate and rapid PoC alternative to RT-PCR.
Collapse
Affiliation(s)
| | - Juliana Breuer
- Lifecare Laboratories, Mainz, Germany
- Pfützner Science and Health Institute, Mainz, Germany
| | - Tuan Ngo Duc
- DHS - Diagnostic HealthCare Solutions, Berlin, Germany
| | - Nicole Thomé
- Lifecare Laboratories, Mainz, Germany
- Pfützner Science and Health Institute, Mainz, Germany
| | | | | | - Julian Kamhieh-Milz
- DHS - Diagnostic HealthCare Solutions, Berlin, Germany.
- Institute of Transfusion Medicine, Charité - Universitätsmedizin Berlin, Robert-Koch Platz 4, 10117, Berlin, Germany.
| | - Andreas Pfützner
- Pfützner Science and Health Institute, Mainz, Germany
- Institute for Internal Medicine and Laboratory Medicine, University for Digital Technologies in Medicine and Dentistry, Wiltz, Luxembourg
| |
Collapse
|
12
|
Lane D, Allsopp R, Holmes CW, Slingsby OC, Jukes-Jones R, Bird P, Anderson NL, Razavi M, Yip R, Pearson TW, Pope M, Khunti K, Doykov I, Hällqvist J, Mills K, Skipp P, Carling R, Ng L, Shaw J, Gupta P, Jones DJL. A high throughput immuno-affinity mass spectrometry method for detection and quantitation of SARS-CoV-2 nucleoprotein in human saliva and its comparison with RT-PCR, RT-LAMP, and lateral flow rapid antigen test. Clin Chem Lab Med 2024; 62:1206-1216. [PMID: 38253336 DOI: 10.1515/cclm-2023-0243] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024]
Abstract
OBJECTIVES Many reverse transcription polymerase chain reaction (RT-PCR) methods exist that can detect SARS-CoV-2 RNA in different matrices. RT-PCR is highly sensitive, although viral RNA may be detected long after active infection has taken place. SARS-CoV-2 proteins have shorter detection windows hence their detection might be more meaningful. Given salivary droplets represent a main source of transmission, we explored the detection of viral RNA and protein using four different detection platforms including SISCAPA peptide immunoaffinity liquid chromatography-mass spectrometry (SISCAPA-LC-MS) using polyclonal capture antibodies. METHODS The SISCAPA-LC MS method was compared to RT-PCR, RT-loop-mediated isothermal amplification (RT-LAMP), and a lateral flow rapid antigen test (RAT) for the detection of virus material in the drool saliva of 102 patients hospitalised after infection with SARS-CoV-2. Cycle thresholds (Ct) of RT-PCR (E gene) were compared to RT-LAMP time-to-positive (TTP) (NE and Orf1a genes), RAT optical densitometry measurements (test line/control line ratio) and to SISCAPA-LC-MS for measurements of viral protein. RESULTS SISCAPA-LC-MS showed low sensitivity (37.7 %) but high specificity (89.8 %). RAT showed lower sensitivity (24.5 %) and high specificity (100 %). RT-LAMP had high sensitivity (83.0 %) and specificity (100.0 %). At high initial viral RNA loads (<20 Ct), results obtained using SISCAPA-LC-MS correlated with RT-PCR (R2 0.57, p-value 0.002). CONCLUSIONS Detection of SARS-CoV-2 nucleoprotein in saliva was less frequent than the detection of viral RNA. The SISCAPA-LC-MS method allowed processing of multiple samples in <150 min and was scalable, enabling high throughput.
Collapse
Affiliation(s)
- Dan Lane
- The Department of Chemical Pathology and Metabolic Diseases, Leicester Royal Infirmary, University Hospitals of Leicester, Leicester, UK
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Rebecca Allsopp
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Christopher W Holmes
- Clinical Microbiology, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, UK
| | | | - Rebekah Jukes-Jones
- The Department of Chemical Pathology and Metabolic Diseases, Leicester Royal Infirmary, University Hospitals of Leicester, Leicester, UK
| | - Paul Bird
- Clinical Microbiology, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, UK
| | | | | | - Richard Yip
- SISCAPA Assay Technologies, Inc., Washington, DC, USA
| | | | - Matt Pope
- SISCAPA Assay Technologies, Inc., Washington, DC, USA
| | - Kamlesh Khunti
- Leicester Diabetes Centre, Leicester General Hospital, University of Leicester, Leicester, UK
| | - Ivan Doykov
- Genetics & Genomic Medicine Department, Translational Mass Spectrometry Research Group, UCL Institute of Child Health, London, UK
- Great Ormond Street Biomedical Research Centre, UCL Institute of Child Health, London, UK
| | - Jenny Hällqvist
- Genetics & Genomic Medicine Department, Translational Mass Spectrometry Research Group, UCL Institute of Child Health, London, UK
- Great Ormond Street Biomedical Research Centre, UCL Institute of Child Health, London, UK
| | - Kevin Mills
- Genetics & Genomic Medicine Department, Translational Mass Spectrometry Research Group, UCL Institute of Child Health, London, UK
- Great Ormond Street Biomedical Research Centre, UCL Institute of Child Health, London, UK
| | - Paul Skipp
- Centre for Proteomic Research, University of Southampton, Southampton, UK
| | - Rachel Carling
- Biochemical Sciences, Synnovis, Guys & St Thomas' NHSFT, London, UK
- GKT School Medical Education, Kings College London, London, UK
| | - Leong Ng
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- van Geest MS-OMICS Facility, University of Leicester, Leicester, UK
| | - Jacqui Shaw
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Pankaj Gupta
- The Department of Chemical Pathology and Metabolic Diseases, Leicester Royal Infirmary, University Hospitals of Leicester, Leicester, UK
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Donald J L Jones
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
- van Geest MS-OMICS Facility, University of Leicester, Leicester, UK
| |
Collapse
|
13
|
Zhou C, Cai Z, Jin B, Lin H, Xu L, Jin Z. Saliva-based detection of SARS-CoV-2: a bibliometric analysis of global research. Mol Cell Biochem 2024; 479:761-777. [PMID: 37178376 PMCID: PMC10182745 DOI: 10.1007/s11010-023-04760-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
Saliva has emerged as a promising noninvasive biofluid for the diagnosis of oral and systemic diseases, including viral infections. During the coronavirus disease 2019 (COVID-19) pandemic, a growing number of studies focused on saliva-based detection of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Taking advantage of the WoS core collection (WoSCC) and CiteSpace, we retrieved 1021 articles related to saliva-based detection of SARS-CoV-2 and conducted a comprehensive bibliometric analysis. We analyzed countries, institutions, authors, cited authors, and cited journals to summarize their contribution and influence and analyzed keywords to explore research hotspots and trends. From 2020 to 2021, research focused on viral transmission via saliva and verification of saliva as a reliable specimen, whereas from 2021 to the present, the focus of research has switched to saliva-based biosensors for SARS-CoV-2 detection. By far, saliva has been verified as a reliable specimen for SARS-CoV-2 detection, although a standardized procedure for saliva sampling and processing is needed. Studies on saliva-based detection of SARS-CoV-2 will promote the development of saliva-based diagnostics and biosensors for viral detection. Collectively, our findings could provide valuable information to help scientists perceive the basic knowledge landscapes on saliva-based detection of SARS-CoV-2, the past and current research hotspots, and future opportunities.
Collapse
Affiliation(s)
- Chun Zhou
- Jinhua People's Hospital Joint Center for Biomedical Research, Zhejiang Normal University, Jinhua, 321000, Zhejiang, China
- Department of Science and Education, the Affiliated Jinhua Hospital of Wenzhou Medical University, Jinhua, 321000, Zhejiang, China
| | - Zhaopin Cai
- College of Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321000, Zhejiang, China
| | - Boxing Jin
- College of Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321000, Zhejiang, China
| | - Huisong Lin
- Zhejiang Institute of Medical Device Testing, Hangzhou, Zhejiang, China
| | - Lingling Xu
- College of Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321000, Zhejiang, China
| | - Zhigang Jin
- Jinhua People's Hospital Joint Center for Biomedical Research, Zhejiang Normal University, Jinhua, 321000, Zhejiang, China.
- College of Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321000, Zhejiang, China.
| |
Collapse
|
14
|
Lei Y, Xu D. Rapid Nucleic Acid Diagnostic Technology for Pandemic Diseases. Molecules 2024; 29:1527. [PMID: 38611806 PMCID: PMC11013254 DOI: 10.3390/molecules29071527] [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: 02/21/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
The recent global pandemic of coronavirus disease 2019 (COVID-19) has enormously promoted the development of diagnostic technology. To control the spread of pandemic diseases and achieve rapid screening of the population, ensuring that patients receive timely treatment, rapid diagnosis has become the top priority in the development of clinical technology. This review article aims to summarize the current rapid nucleic acid diagnostic technologies applied to pandemic disease diagnosis, from rapid extraction and rapid amplification to rapid detection. We also discuss future prospects in the development of rapid nucleic acid diagnostic technologies.
Collapse
Affiliation(s)
- Yu Lei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Chinese Academy of Sciences (CAS), Beijing 100190, China;
- University of Chinese Academy of Sciences (UCAS), Beijing 100049, China
| | - Dawei Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Chinese Academy of Sciences (CAS), Beijing 100190, China;
| |
Collapse
|
15
|
Alsaeed M, Alhamid G, Tombuloglu H, Kabanja JH, Karagoz A, Tombuloglu G, Rabaan AA, Al-Suhaimi E, Unver T. Ultrasensitive and fast detection of SARS-CoV-2 using RT-LAMP without pH-dependent dye. Funct Integr Genomics 2024; 24:16. [PMID: 38242999 DOI: 10.1007/s10142-024-01297-z] [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: 11/28/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/21/2024]
Abstract
This study investigates the performance of reverse transcription loop-mediated isothermal amplification (RT-LAMP) assay for the colorimetric detection of SARS-CoV-2 using fluorometric dye, namely, calcein. The detection limit (LoD) with the N-ID1 primer set resulted in superior performance, corresponding to ~ 2 copies/reaction or ~ 0.1 copies/μL of the RNA sample. The color development can be observed by the naked eye, using an ultraviolet (UV) transilluminator or a hand-UV light without the requirement of expensive devices. The average time-to-reaction (TTR) value was 26.2 min in high-copy number samples, while it was about 50 min in rRT-PCR. A mobile application was proposed to quantify the positive and negative results based on the three-color spaces (RGB, Lab, and HSB). Compared to rRT-PCR (n = 67), this assay allows fast and sensitive visual detection of SARS-CoV-2, with high sensitivity (90.9%), selectivity (100%), and accuracy (94.03%). Besides, the assay was sensitive regardless of variants. Since this assay uses a fluorescent dye for visual observation, it can be easily adapted in RT-LAMP assays with high sensitivity. Thus, it can be utilized in low-source centers and field testing such as conferences, sports meetings, refugee camps, companies, and schools.
Collapse
Affiliation(s)
- Moneerah Alsaeed
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Galyah Alhamid
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Huseyin Tombuloglu
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia.
| | - Juma H Kabanja
- Department of Pathology & Laboratory Medicine, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Aysel Karagoz
- Quality Assurance Department, Turk Pharmaceutical and Serum Ind. Inc., Ankara, Turkey
| | - Guzin Tombuloglu
- Department of Biophysics, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan
| | - Ebtesam Al-Suhaimi
- Vice Presidency for Scientific Research and Innovation, Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - Turgay Unver
- Ficus Biotechnology, Ankara, Turkey
- Faculty of Engineering, Ostim Technical University, 06374, Ankara, Turkey
| |
Collapse
|
16
|
Uddin MKM, Hossain ME, Ami JQ, Hasan R, Hasan MM, Islam A, Hasan MJ, Shaly NJ, Ahmed S, Samina P, Rahman MZ, Rahman M, Banu S. SalivaDirect: an alternative to a conventional RNA extraction protocol for molecular detection of SARS-CoV-2 in a clinical setting. Microbiol Spectr 2024; 12:e0327223. [PMID: 38014980 PMCID: PMC10783095 DOI: 10.1128/spectrum.03272-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/01/2023] [Indexed: 11/29/2023] Open
Abstract
IMPORTANCE Affordable and accessible tests for COVID-19 allow for timely disease treatment and pandemic management. SalivaDirect is a faster and easier method to implement than NPS sampling. Patients can self-collect saliva samples at home or in other non-clinical settings without the help of a healthcare professional. Sample processing in SalivaDirect is less complex and more adaptable than in conventional nucleic acid extraction methods. We found that SalivaDirect has good diagnostic performance and is ideal for large-scale testing in settings where supplies may be limited or trained healthcare professionals are unavailable.
Collapse
Affiliation(s)
| | - Mohammad Enayet Hossain
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Jenifar Quaiyum Ami
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Rashedul Hasan
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Md. Mahmudul Hasan
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Ashabul Islam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Md. Jahid Hasan
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Nusrat Jahan Shaly
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Shahriar Ahmed
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Pushpita Samina
- Centre for Health Economics and Policy Analysis, McMaster University, Hamilton, Ontario, Canada
| | - Mohammed Ziaur Rahman
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Mustafizur Rahman
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Sayera Banu
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| |
Collapse
|
17
|
Ebbah E, Amissah A, Kim JH, Driskell JD. SERS-based immunoassay on a plasmonic syringe filter for improved sampling and labeling efficiency of biomarkers. Analyst 2023; 149:221-230. [PMID: 38018888 DOI: 10.1039/d3an01899g] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Rapid, sensitive, and quantitative detection of biomarkers is needed for early diagnosis of disease and surveillance of infectious outbreaks. Here, we exploit a plasmonic syringe filter and surface-enhanced Raman spectroscopy (SERS) in the development of a rapid detection system, using human IgG as a model diagnostic biomarker. The novel assay design facilitates multiple passages of the sample and labeling solution through the detection zone enabling us to investigate and maximize sampling efficiency to the capture substrate. The vertical flow immunoassay process in this study involves the utilization of filter paper embedded with gold nanoparticles (AuNPs) to form a plasmonic substrate. Capture antibody (anti-human IgG) is then immobilized onto the prepared plasmonic paper and inserted into a vertical flow device (syringe filter holder). Sample solution is passed through the filter paper and the target antigen (human IgG) is selectively captured by the immobilized antibody to form an antibody-antigen complex. Next, functionalized AuNPs as extrinsic Raman labels (ERLs) are passed through the filter paper to label the captured biomarker molecules forming a layered structure. This sandwiched geometry enhances plasmonic coupling and SERS signal to provide highly sensitive detection of biomolecules. Systematic studies to investigate the impact of multiple infuse/withdraw cycles of the sample and labeling solutions reveal that antigen and ERL binding are maximized with 10 and 20 cycles, respectively. The optimized assay achieves a detection limit of ∼0.2 ng mL-1 for human IgG with a total assay time of less than 5 minutes, meeting the demands for rapid point of care diagnostics. Additionally, the optimized platform was implemented in the quantitative analysis of the SARS-CoV-2 nucleocapsid protein, the typical target in commercial, FDA-approved rapid antigen tests for COVID-19.
Collapse
Affiliation(s)
- Eunice Ebbah
- Department of Chemistry, Illinois State University, Normal, IL 61790, USA.
| | - Anthony Amissah
- Department of Chemistry, Illinois State University, Normal, IL 61790, USA.
| | - Jun-Hyun Kim
- Department of Chemistry, Illinois State University, Normal, IL 61790, USA.
| | - Jeremy D Driskell
- Department of Chemistry, Illinois State University, Normal, IL 61790, USA.
| |
Collapse
|
18
|
Wang W, Shi S, Liu Y, Hou Z, Qi J, Guo L. Staging classification of omicron variant SARS-CoV-2 infection based on dual-spectrometer LIBS (DS-LIBS) combined with machine learning. OPTICS EXPRESS 2023; 31:42413-42427. [PMID: 38087616 DOI: 10.1364/oe.504640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/15/2023] [Indexed: 12/18/2023]
Abstract
Effective differentiation of the infection stages of omicron can provide significant assistance in transmission control and treatment strategies. The combination of LIBS serum detection and machine learning methods, as a novel disease auxiliary diagnostic approach, has a high potential for rapid and accurate staging classification of Omicron infection. However, conventional single-spectrometer LIBS serum detection methods focus on detecting the spectra of major elements, while trace elements are more closely related to the progression of COVID-19. Here, we proposed a rapid analytical method with dual-spectrometer LIBS (DS-LIBS) assisted with machine learning to classify different infection stages of omicron. The DS-LIBS, including a broadband spectrometer and a narrowband spectrometer, enables synchronous collection of major and trace elemental spectra in serum, respectively. By employing the RF machine learning models, the classification accuracy using the spectra data collected from DS-LIBS can reach 0.92, compared to 0.84 and 0.73 when using spectra data collected from single-spectrometer LIBS. This significant improvement in classification accuracy highlights the efficacy of the DS-LIBS approach. Then, the performance of four different models, SVM, RF, IGBT, and ETree, is compared. ETree demonstrates the best, with cross-validation and test set accuracies of 0.94 and 0.93, respectively. Additionally, it achieves classification accuracies of 1.00, 0.92, 0.92, and 0.89 for the four stages B1-acute, B1-post, B2, and B3. Overall, the results demonstrate that DS-LIBS combined with the ETree machine learning model enables effective staging classification of omicron infection.
Collapse
|
19
|
Lin Z, Sun B, Yang X, Jiang Y, Wu S, Lv B, Pan Y, Zhang Q, Wang X, Xiang G, Lou Y, Xiao X. Infectious Disease Diagnosis and Pathogen Identification Platform Based on Multiplex Recombinase Polymerase Amplification-Assisted CRISPR-Cas12a System. ACS Infect Dis 2023; 9:2306-2315. [PMID: 37811564 DOI: 10.1021/acsinfecdis.3c00381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Controlling and mitigating infectious diseases caused by multiple pathogens or pathogens with several subtypes require multiplex nucleic acid detection platforms that can detect several target genes rapidly, specifically, sensitively, and simultaneously. Here, we develop a detection platform, termed Multiplex Assay of RPA and Collateral Effect of Cas12a-based System (MARPLES), based on multiplex nucleic acid amplification and Cas12a ssDNase activation to diagnose these diseases and identify their pathogens. We use the clinical specimens of hand, foot, and mouth disease (HFMD) and influenza A to evaluate the feasibility of MARPLES in diagnosing the disease and identifying the pathogen, respectively, and find that MARPLES can accurately diagnose the HFMD associated with enterovirus 71, coxsackievirus A16 (CVA16), CVA6, or CVA10 and identify the exact types of H1N1 and H3N2 in an hour, showing high sensitivity and specificity and 100% predictive agreement with qRT-PCR. Collectively, our findings demonstrate that MARPLES is a promising multiplex nucleic acid detection platform for disease diagnosis and pathogen identification.
Collapse
Affiliation(s)
- Ziqin Lin
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Baochang Sun
- Department of Laboratory, Wenzhou Center for Disease Control and Prevention, Wenzhou 325035, China
| | - Xi Yang
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yayun Jiang
- Department of Clinical Laboratory, People's Hospital of Deyang City, Deyang 618000, China
| | - Sihong Wu
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Binbin Lv
- Department of Laboratory, Wenzhou Center for Disease Control and Prevention, Wenzhou 325035, China
| | - Yajing Pan
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qingxun Zhang
- Beijing Milu Ecological Research Center, Beijing Academy of Science and Technology, Beijing 100076, China
| | - Xiaoqiong Wang
- Zhuji Institute of Biomedicine, Wenzhou Medical University, Zhuji, Shaoxing 311800, Zhejiang, China
| | - Guangxin Xiang
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yongliang Lou
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xingxing Xiao
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
20
|
Wang Q, Panpradist N, Kotnik JH, Willson RC, Kourentzi K, Chau ZL, Liu JK, Lutz BR, Lai JJ. A simple agglutination system for rapid antigen detection from large sample volumes with enhanced sensitivity. Anal Chim Acta 2023; 1277:341674. [PMID: 37604625 PMCID: PMC10777812 DOI: 10.1016/j.aca.2023.341674] [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: 04/27/2023] [Revised: 07/14/2023] [Accepted: 07/29/2023] [Indexed: 08/23/2023]
Abstract
Lateral flow assays (LFAs) provide a simple and quick option for diagnosis and are widely adopted for point-of-care or at-home tests. However, their sensitivity is often limited. Most LFAs only allow 50 μL samples while various sample types such as saliva could be collected in much larger volumes. Adapting LFAs to accommodate larger sample volumes can improve assay sensitivity by increasing the number of target analytes available for detection. Here, a simple agglutination system comprising biotinylated antibody (Ab) and streptavidin (SA) is presented. The Ab and SA agglutinate into large aggregates due to multiple biotins per Ab and multiple biotin binding sites per SA. Dynamic light scattering (DLS) measurements showed that the agglutinated aggregate could reach a diameter of over 0.5 μm and over 1.5 μm using poly-SA. Through both experiments and Monte Carlo modeling, we found that high valency and equivalent concentrations of the two aggregating components were critical for successful agglutination. The simple agglutination system enables antigen capture from large sample volumes with biotinylated Ab and a swift transition into aggregates that can be collected via filtration. Combining the agglutination system with conventional immunoassays, an agglutination assay is proposed that enables antigen detection from large sample volumes using an in-house 3D-printed device. As a proof-of-concept, we developed an agglutination assay targeting SARS-CoV-2 nucleocapsid antigen for COVID-19 diagnosis from saliva. The assay showed a 10-fold sensitivity enhancement when increasing sample volume from 50 μL to 2 mL, with a final limit of detection (LoD) of 10 pg mL-1 (∼250 fM). The assay was further validated in negative saliva spiked with gamma-irradiated SARS-CoV-2 and showed an LoD of 250 genome copies per μL. The proposed agglutination assay can be easily developed from existing LFAs to facilitate the processing of large sample volumes for improved sensitivity.
Collapse
Affiliation(s)
- Qin Wang
- Department of Bioengineering, University of Washington, Seattle, WA, 98195-5061, USA
| | - Nuttada Panpradist
- Department of Bioengineering, University of Washington, Seattle, WA, 98195-5061, USA
| | - Jack Henry Kotnik
- Department of Bioengineering, University of Washington, Seattle, WA, 98195-5061, USA
| | - Richard C Willson
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, 77204, USA
| | - Katerina Kourentzi
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, 77204, USA
| | - Zoe L Chau
- Department of Bioengineering, University of Washington, Seattle, WA, 98195-5061, USA
| | - Joanne K Liu
- Department of Bioengineering, University of Washington, Seattle, WA, 98195-5061, USA
| | - Barry R Lutz
- Department of Bioengineering, University of Washington, Seattle, WA, 98195-5061, USA.
| | - James J Lai
- Department of Bioengineering, University of Washington, Seattle, WA, 98195-5061, USA; Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, 106335, Taiwan.
| |
Collapse
|
21
|
Daum LT, Rodriguez JD, Ward SR, Chambers JP. Extraction-Free Detection of SARS-CoV-2 Viral RNA Using LumiraDx's RNA Star Complete Assay from Clinical Nasal Swabs Stored in a Novel Collection and Transport Medium. Diagnostics (Basel) 2023; 13:3010. [PMID: 37761377 PMCID: PMC10528020 DOI: 10.3390/diagnostics13183010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Background: The rapid detection of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is vital for patient care. The LumiraDx™ SARS-CoV-2 RNA Star Complete (RSC) is an Emergency Use Authorization-recognized molecular test using nasal/nasopharyngeal swabs immersed in a viral/universal transport medium (VTM/UTM). However, there is a critical need for an alternative medium for point-of-care testing (POCT). This study aimed to investigate Xtract-Free (XF), a novel collection medium for transport and direct (extraction-free) use with nucleic acid tests. Methods: Using serially diluted SARS-CoV-2 viral RNA (vRNA) in a routine UTM and XF, a limit of detection (LOD) was established via an RSC test and a quantitative reverse transcription PCR (RT-qPCR). Additionally, the results obtained from a panel of 108 clinical "car-side" nasal swabs collected in XF during the coronavirus pandemic and assessed using the "gold-standard" RT-qPCR assay were compared to Lumira's RSC assay. Results: The average replicate RT-qPCR cycle threshold (CT) values for vRNA in XF and UTM were observed to be equivalent. An LOD for which five out of five replicates were detected using XF or VTM was approximately 2000 copies/mL. The nasal swabs collected in XF exhibited 93.9% positive percent agreement (sensitivity) and 100% negative percent agreement (specificity) compared to the RT-qPCR. Three specimens tested positive via an RT-qPCR were negative when tested via RSC; however, all three samples had CT values ≥ 36.4. Conclusions: XF is equivalent to VTM/UTM and is compatible for use with the RSC test. Furthermore, XF can be used directly with RT-qPCRs and rapid antigen testing without the requirement for separate nucleic acid extraction (an extraction-free process), making it ideal for cost-effective high-throughput and decentralized respiratory testing. Impact Statement: This study is the first to evaluate LumiraDx's SARS-CoV-2 RNA Star Complete assay in concert with Xtract-Free (XF), a novel collection medium containing a proprietary RNase-inactivating technology for the rapid, "extraction-free" detection of SARS-CoV-2 RNA from clinical nasal swabs. Specimens collected in XF combined with rapid LumiraDx detection provide a safe and sensitive alternative to VTM/UTM, and Molecular Transport medium (MTM) for high throughput, "extraction-free" molecular detection.
Collapse
Affiliation(s)
- Luke T. Daum
- LuJo BioScience Laboratory, 1747 Citadel Plaza, Suite 201, San Antonio, TX 78209, USA;
| | - John D. Rodriguez
- LuJo BioScience Laboratory, 1747 Citadel Plaza, Suite 201, San Antonio, TX 78209, USA;
| | | | - James P. Chambers
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA;
| |
Collapse
|
22
|
Tayyab M, Barrett D, van Riel G, Liu S, Reinius B, Scharfe C, Griffin P, Steinmetz LM, Javanmard M, Pelechano V. Digital assay for rapid electronic quantification of clinical pathogens using DNA nanoballs. SCIENCE ADVANCES 2023; 9:eadi4997. [PMID: 37672583 PMCID: PMC10482329 DOI: 10.1126/sciadv.adi4997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/04/2023] [Indexed: 09/08/2023]
Abstract
Fast and accurate detection of nucleic acids is key for pathogen identification. Methods for DNA detection generally rely on fluorescent or colorimetric readout. The development of label-free assays decreases costs and test complexity. We present a novel method combining a one-pot isothermal generation of DNA nanoballs with their detection by electrical impedance. We modified loop-mediated isothermal amplification by using compaction oligonucleotides that self-assemble the amplified target into nanoballs. Next, we use capillary-driven flow to passively pass these nanoballs through a microfluidic impedance cytometer, thus enabling a fully compact system with no moving parts. The movement of individual nanoballs is detected by a change in impedance providing a quantized readout. This approach is flexible for the detection of DNA/RNA of numerous targets (severe acute respiratory syndrome coronavirus 2, HIV, β-lactamase gene, etc.), and we anticipate that its integration into a standalone device would provide an inexpensive (<$5), sensitive (10 target copies), and rapid test (<1 hour).
Collapse
Affiliation(s)
- Muhammad Tayyab
- Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Donal Barrett
- SciLifeLab, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Gijs van Riel
- SciLifeLab, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Shujing Liu
- SciLifeLab, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
- International Institute of Tea Industry Innovation for the Belt and Road, Nanjing Agricultural University, Nanjing 210095, China
| | - Björn Reinius
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | | | - Peter Griffin
- Stanford Genome Technology Center, Stanford, CA, USA
| | - Lars M. Steinmetz
- Stanford Genome Technology Center, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Mehdi Javanmard
- Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Vicent Pelechano
- SciLifeLab, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
23
|
Kubo T, Kanao E, Ishida K, Minami S, Tanigawa T, Mizuta R, Sasaki Y, Otsuka K, Kobayashi T. Efficient Selective Adsorption of SARS-CoV-2 via the Recognition of Spike Proteins Using an Affinity Spongy Monolith. Anal Chem 2023; 95:13185-13190. [PMID: 37610704 DOI: 10.1021/acs.analchem.3c02097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Since the outbreak of COVID-19, SARS-CoV-2, the infection has been spreading to date. The rate of false-negative result on a polymerase chain reaction (PCR) test considered the gold standard is roughly 20%. Therefore, its accuracy poses a question as well as needs improvement in the test. This study reports fabrication of a substrate of an anti-spike protein (AS)-immobilized porous material having selective adsorption toward a spike protein protruding from the surface of SARS-CoV-2. We have employed an organic polymer substrate called spongy monolith (SPM). The SPM has through-pores of about 10 μm and is adequate for flowing liquid containing virus particles. It also involves an epoxy group on the surface, enabling arbitrary proteins such as antibodies to immobilize. When antibodies of the spike protein toward receptor binding domain were immobilized, selective adsorption of the spike protein was observed. At the same time, when mixed analytes of spike proteins, lysozymes and amylases, were flowed into an AS-SPM, selective adsorption toward the spike proteins was observed. Then, SARS-CoV-2 was flowed into the BSA-SPM or AS-SPM, amounts of SARS-CoV-2 adsorption toward the AS-SPM were much larger compared to the ones toward the BSA-SPM. Furthermore, rotavirus was not adsorbed to the AS-SPM at all. These results show that the AS-SPM recognizes selectively the spike proteins of SARS-CoV-2 and may be possible applications for the purification and concentration of SARS-CoV-2.
Collapse
Affiliation(s)
- Takuya Kubo
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Eisuke Kanao
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Ibaraki 567-0085, Japan
| | - Koki Ishida
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Shohei Minami
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Tetsuya Tanigawa
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Ryosuke Mizuta
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-Ko, Kyoto 615-8510, Japan
| | - Yoshihiro Sasaki
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-Ko, Kyoto 615-8510, Japan
| | - Koji Otsuka
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Takeshi Kobayashi
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
24
|
Baek YH, Park MY, Lim HJ, Youm DJ, You Y, Ahn S, Park JE, Kim MJ, Lee SH, Sohn YH, Yang YJ. Evaluation of Rapid Multiplex Reverse Transcription-Quantitative Polymerase Chain Reaction Assays for SARS-CoV-2 Detection in Individual and Pooled Samples. Life (Basel) 2023; 13:1717. [PMID: 37629574 PMCID: PMC10455980 DOI: 10.3390/life13081717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Although coronavirus disease 2019 (COVID-19) is no longer a Public Health Emergency of International Concern (PHEIC), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has had a vast impact to date. Hence, continuous management is required, given the uncertainty caused by the potential evolution of SARS-CoV-2. Reverse transcription-quantitative PCR (RT-qPCR) diagnosis has been fundamental in overcoming this issue. In this study, the performances of two rapid RT-qPCR assays (Real-Q Direct SARS-CoV-2 Detection Kit and Allplex™ SARS-CoV-2 fast PCR Assay) with short PCR times were comparatively evaluated using a STANDARD M nCoV Real-Time Detection Kit (STANDARD M, conventional RT-qPCR assay). All kits showed a limit of detection values (102-103 copies/reaction). The evaluation showed that the two rapid assay tests had ≥97.89% sensitivity and ≥99.51% specificity (κ = 0.98) for individual samples and ≥97.32% sensitivity and ≥97.67% specificity for pooled samples compared to STANDARD M. These results indicate that the two rapid RT-qPCR kits, which showed significant time reduction in performance, are as effective as a conventional RT-qPCR assay. They are likely to increase not only the number of tests that can be performed but also the efficiency of sustainable management of COVID-19 in the long term.
Collapse
Affiliation(s)
- Young-Hyun Baek
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Min-Young Park
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Ho-Jae Lim
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea;
| | - Dong-Jae Youm
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Youngshin You
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Seojin Ahn
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Jung-Eun Park
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea;
| | - Min-Jin Kim
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Sun-Hwa Lee
- Department of Laboratory Medicine, Seegene Medical Foundation, Seoul 04805, Republic of Korea;
| | - Yong-Hak Sohn
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Yong-Jin Yang
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| |
Collapse
|
25
|
Reyes-Morales R, Segundo-Ibañez P, Flores-de Los Ángeles C, Vizcarra-Ramos D, Ibañez-Galeana DI, Salas-Cuevas G, Olvera-Serrano Á, Pérez-Silva NB, Rocha-Rocha VM, El-Kassi EG, Escobedo-Straffon J, Contreras-Mioni L, Rosas-Díaz M, Lopez-Martinez KM, Arias-Matus CE, Bautista-Rodriguez E, Nolasco-Quiroga M. Reverse transcription loop‑mediated isothermal amplification has a high performance in the detection of SARS‑CoV‑2 in saliva samples and nasal swabs from asymptomatic and symptomatic individuals. Exp Ther Med 2023; 26:398. [PMID: 37522063 PMCID: PMC10375439 DOI: 10.3892/etm.2023.12097] [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: 01/10/2023] [Accepted: 05/31/2023] [Indexed: 08/01/2023] Open
Abstract
The detection of coronavirus disease 2019 cases represents a significant challenge at the epidemiological level. Limitations exist in effectively detecting asymptomatic cases, achieving good follow-up in hospitals without the infrastructure for reverse transcription-quantitative PCR (RT-qPCR) or in difficult-to-access areas and developing methods with the need for less invasive sampling procedures. Therefore, the present study evaluated the performance of the direct reverse transcription loop-mediated isothermal amplification (RT-LAMP) test for detecting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the saliva and nasal samples of asymptomatic individuals belonging to the university population. In addition, this test was also assessed for effectiveness in symptomatic individuals referred from a hospital with poor infrastructure in molecular biology and located outside the urban area. The RT-LAMP assay was compared with the results obtained from the RT-qPCR nasopharyngeal swab test, where the diagnosis was confirmed by lateral flow immunoassay test for rapid antigen detection. A total of 128 samples were analyzed, of which 43% were symptomatic positive individuals, 25% were asymptomatic positive individuals and 32% were SARS-CoV2-negative control individuals. Among positive individuals, no differences were found between the Cq values determined by RT-qPCR. A sensitivity of 96.5% and a specificity of 97.6% was reported for the detection of SARS-CoV-2 in symptomatic individuals by salivary and nasal RT-LAMP, as well as a sensitivity of 100% and a specificity of 97.6% for the detection of SARS-CoV-2 in asymptomatic individuals. These findings indicated that performance of the direct RT-LAMP test using saliva and nasal samples has high sensitivity and specificity, which in turn suggest that it is a viable and reliable alternative for use in epidemiological monitoring.
Collapse
Affiliation(s)
- Rodolfo Reyes-Morales
- Laboratory of Medical and Pharmaceutical Biotechnology, Biotechnology Faculty, Puebla State Popular Autonomous University, Puebla 72410, Mexico
| | - Patricia Segundo-Ibañez
- Molecular Biology Laboratory, Biotechnology Department, Interamerican University, Puebla 72828, Mexico
| | - César Flores-de Los Ángeles
- Molecular Diagnostic Laboratory, Biotechnology Faculty, Puebla State Popular Autonomous University, Puebla 72410, Mexico
| | - David Vizcarra-Ramos
- Molecular Biology Laboratory, Biotechnology Department, Interamerican University, Puebla 72828, Mexico
| | | | - Gabriela Salas-Cuevas
- COVID Area of Hospital Clinic Huauchinango, Institute of Social Security and Services for State Workers, Huauchinango, Puebla 73160, Mexico
| | - Ángel Olvera-Serrano
- COVID Area of Hospital Clinic Huauchinango, Institute of Social Security and Services for State Workers, Huauchinango, Puebla 73160, Mexico
| | - Nancy Bibiana Pérez-Silva
- Molecular Diagnostic Laboratory, Biotechnology Faculty, Puebla State Popular Autonomous University, Puebla 72410, Mexico
| | - Valeria Magali Rocha-Rocha
- Biological Science Department, Biotechnology Faculty, Puebla State Popular Autonomous University, Puebla 72410, Mexico
| | - Elie Girgis El-Kassi
- Biological Science Department, Biotechnology Faculty, Puebla State Popular Autonomous University, Puebla 72410, Mexico
| | - Jorge Escobedo-Straffon
- Biological Science Department, Biotechnology Faculty, Puebla State Popular Autonomous University, Puebla 72410, Mexico
| | - Laura Contreras-Mioni
- Biological Science Department, Biotechnology Faculty, Puebla State Popular Autonomous University, Puebla 72410, Mexico
| | - Marisol Rosas-Díaz
- Molecular Biology Laboratory, Multidisciplinary Academic Unit Reynosa-Aztlan Reynosa, Autonomous University of Tamaulipas, Tamaulipas 88740, Mexico
| | - Karla María Lopez-Martinez
- Laboratory of Medical and Pharmaceutical Biotechnology, Biotechnology Faculty, Puebla State Popular Autonomous University, Puebla 72410, Mexico
| | - Carlos Eduardo Arias-Matus
- Laboratory of Medical and Pharmaceutical Biotechnology, Biotechnology Faculty, Puebla State Popular Autonomous University, Puebla 72410, Mexico
| | - Elizabeth Bautista-Rodriguez
- Laboratory of Medical and Pharmaceutical Biotechnology, Biotechnology Faculty, Puebla State Popular Autonomous University, Puebla 72410, Mexico
| | - Manuel Nolasco-Quiroga
- COVID Area of Hospital Clinic Huauchinango, Institute of Social Security and Services for State Workers, Huauchinango, Puebla 73160, Mexico
| |
Collapse
|
26
|
Arai Y, Takahashi K, Horinouchi T, Takahashi K, Ozaki H. SAGAS: Simulated annealing and greedy algorithm scheduler for laboratory automation. SLAS Technol 2023; 28:264-277. [PMID: 36997066 DOI: 10.1016/j.slast.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/07/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
During laboratory automation of life science experiments, coordinating specialized instruments and human experimenters for various experimental procedures is important to minimize the execution time. In particular, the scheduling of life science experiments requires the consideration of time constraints by mutual boundaries (TCMB) and can be formulated as the "scheduling for laboratory automation in biology" (S-LAB) problem. However, existing scheduling methods for the S-LAB problems have difficulties in obtaining a feasible solution for large-size scheduling problems at a time sufficient for real-time use. In this study, we proposed a fast schedule-finding method for S-LAB problems, SAGAS (Simulated annealing and greedy algorithm scheduler). SAGAS combines simulated annealing and the greedy algorithm to find a scheduling solution with the shortest possible execution time. We have performed scheduling on real experimental protocols and shown that SAGAS can search for feasible or optimal solutions in practicable computation time for various S-LAB problems. Furthermore, the reduced computation time by SAGAS enables us to systematically search for laboratory automation with minimum execution time by simulating scheduling for various laboratory configurations. This study provides a convenient scheduling method for life science automation laboratories and presents a new possibility for designing laboratory configurations.
Collapse
Affiliation(s)
- Yuya Arai
- College of Biological Sciences, School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; Bioinformatics Laboratory, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Ko Takahashi
- College of Biological Sciences, School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Takaaki Horinouchi
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo, 135-0064, Japan; Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, 6-2-3 Furuedai, Suita, Osaka, 565-0874, Japan
| | - Koichi Takahashi
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, 6-2-3 Furuedai, Suita, Osaka, 565-0874, Japan; Graduate School of Media and Governance, Keio University, 5322 Endo, Fujisawa, Kanagawa, 252-0816, Japan
| | - Haruka Ozaki
- Bioinformatics Laboratory, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; Center for Artificial Intelligence Research, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
27
|
Vaidyanathan S, Wijerathne H, Gamage SST, Shiri F, Zhao Z, Choi J, Park S, Witek MA, McKinney C, Verber M, Hall AR, Childers K, McNickle T, Mog S, Yeh E, Godwin AK, Soper SA. High Sensitivity Extended Nano-Coulter Counter for Detection of Viral Particles and Extracellular Vesicles. Anal Chem 2023; 95:9892-9900. [PMID: 37336762 PMCID: PMC11015478 DOI: 10.1021/acs.analchem.3c00855] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
We present a chip-based extended nano-Coulter counter (XnCC) that can detect nanoparticles affinity-selected from biological samples with low concentration limit-of-detection that surpasses existing resistive pulse sensors by 2-3 orders of magnitude. The XnCC was engineered to contain 5 in-plane pores each with an effective diameter of 350 nm placed in parallel and can provide high detection efficiency for single particles translocating both hydrodynamically and electrokinetically through these pores. The XnCC was fabricated in cyclic olefin polymer (COP) via nanoinjection molding to allow for high-scale production. The concentration limit-of-detection of the XnCC was 5.5 × 103 particles/mL, which was a 1,100-fold improvement compared to a single in-plane pore device. The application examples of the XnCC included counting affinity selected SARS-CoV-2 viral particles from saliva samples using an aptamer and pillared microchip; the selection/XnCC assay could distinguish the COVID-19(+) saliva samples from those that were COVID-19(-). In the second example, ovarian cancer extracellular vesicles (EVs) were affinity selected using a pillared chip modified with a MUC16 monoclonal antibody. The affinity selection chip coupled with the XnCC was successful in discriminating between patients with high grade serous ovarian cancer and healthy donors using blood plasma as the input sample.
Collapse
Affiliation(s)
- Swarnagowri Vaidyanathan
- Bioengineering Program, The University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Harshani Wijerathne
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Sachindra S T Gamage
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Farhad Shiri
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Zheng Zhao
- Bioengineering Program, The University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Junseo Choi
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
- Mechanical & Industrial Engineering Department, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Sunggook Park
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
- Mechanical & Industrial Engineering Department, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Małgorzata A Witek
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Collin McKinney
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27514, United States
| | - Matthew Verber
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27514, United States
| | - Adam R Hall
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
- Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston Salem, North Carolina 27101, United States
| | - Katie Childers
- Bioengineering Program, The University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Taryn McNickle
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Shalee Mog
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Elaine Yeh
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Andrew K Godwin
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
- KU Comprehensive Cancer Center, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Steven A Soper
- Bioengineering Program, The University of Kansas, Lawrence, Kansas 66045, United States
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
- KU Comprehensive Cancer Center, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
- Department of Mechanical Engineering, The University of Kansas, Lawrence, Kansas 66045, United States
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| |
Collapse
|
28
|
Terada N, Akashi Y, Takeuchi Y, Ueda A, Notake S, Nakamura K, Suzuki H. Prospective study of three saliva qualitative antigen testing kits for the detection of SARS-CoV-2 among mainly symptomatic patients in Japan. J Infect Chemother 2023; 29:654-659. [PMID: 36894015 PMCID: PMC9990886 DOI: 10.1016/j.jiac.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/25/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023]
Abstract
INTRODUCTION Rapid qualitative antigen testing has been widely used for the laboratory diagnosis of COVID-19 with nasopharyngeal samples. Saliva samples have been used as alternative samples, but the analytical performance of those samples for qualitative antigen testing has not been sufficiently evaluated. METHODS A prospective observational study evaluated the analytical performance of three In Vitro Diagnostics (IVD) approved COVID-19 rapid antigen detection kits for saliva between June 2022 and July 2022 in Japan using real-time reverse transcription polymerase chain reaction (RT-qPCR) as a reference. A nasopharyngeal sample and a saliva sample were simultaneously obtained, and RT-qPCR was performed. RESULTS In total, saliva samples and nasopharyngeal samples were collected from 471 individuals (RT-qPCR-positive, n = 145) for the analysis. Of these, 96.6% were symptomatic. The median copy numbers were 1.7 × 106 copies/mL for saliva samples and 1.2 × 108 copies/mL for nasopharyngeal samples (p < 0.001). Compared with the reference, the sensitivity and specificity were 44.8% and 99.7% for ImunoAce SARS-CoV-2 Saliva, 57.2% and 99.1% for Espline SARS-CoV-2 N, and 60.0% and 99.1% for QuickChaser Auto SARS-CoV-2, respectively. The sensitivities of all antigen testing kit were 100% for saliva samples with a high viral load (>107 copies/mL), whereas the sensitivities were <70% for high-viral-load nasopharyngeal samples (>107 copies/mL). CONCLUSION COVID-19 rapid antigen detection kits with saliva showed high specificity, but the sensitivity varied among kits, and were also insufficient for the detection of symptomatic COVID-19 patients.
Collapse
Affiliation(s)
- Norihiko Terada
- Department of Infectious Diseases, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; Division of Infectious Diseases, Department of Medicine, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 305-8558, Japan.
| | - Yusaku Akashi
- Department of Infectious Diseases, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; Division of Infectious Diseases, Department of Medicine, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 305-8558, Japan; Akashi Internal Medicine Clinic, 3-1-63 Asahigaoka, Kashiwara, Osaka, 582-0026, Japan.
| | - Yuto Takeuchi
- Division of Infectious Diseases, Department of Medicine, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 305-8558, Japan; Department of Infectious Diseases, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 305-8576, Japan.
| | - Atsuo Ueda
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 305-8558, Japan.
| | - Shigeyuki Notake
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 305-8558, Japan.
| | - Koji Nakamura
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 305-8558, Japan.
| | - Hiromichi Suzuki
- Department of Infectious Diseases, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; Division of Infectious Diseases, Department of Medicine, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 305-8558, Japan; Department of Infectious Diseases, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 305-8576, Japan.
| |
Collapse
|
29
|
Ahti J, Österback R, Keskitalo A, Mokkala K, Vidbäck S, Veikkolainen V, Vuorinen T, Peltola V, Hakanen AJ, Waris M, Laine M. Diagnostic Performance and Tolerability of Saliva and Nasopharyngeal Swab Specimens in the Detection of SARS-CoV-2 by RT-PCR. Microbiol Spectr 2023; 11:e0532422. [PMID: 37093085 PMCID: PMC10269602 DOI: 10.1128/spectrum.05324-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023] Open
Abstract
Saliva is a promising alternative for a nasopharyngeal swab (NPS) in specimen collection to detect SARS-CoV-2. We compared the diagnostic performance and tolerability of saliva collection versus NPS in a clinical setting. Paired NPS and saliva specimens were collected sequentially from participants (n = 250) at the Turku University Hospital drive-in coronavirus testing station in the spring of 2022, with Omicron BA.2 as the dominant SARS-CoV-2 variant. Discomfort and preference for the sampling method were assessed. The specimens were analyzed for SARS-CoV-2 using real-time multiplex reverse transcriptase PCR (RT-PCR) with a laboratory-developed test (LDT) and two commercial kits (PerkinElmer SARS-CoV-2 and PerkinElmer SARS-CoV-2 Plus) for several target genes. Among the 250 participants, 246 had respiratory symptoms. With LDT, SARS-CoV-2 was detected in 135 and 134 participants from NPS and saliva, respectively. Of the 250 specimens, 11 gave a discordant outcome, resulting in excellent agreement between the specimen types (Cohen's kappa coefficient of 0.911; P = 0.763). The cycle threshold (CT) values of LDT and commercial kit target genes were significantly lower from NPS than from saliva. A total of 172 (69%) participants assessed saliva sampling as more tolerable than NPS (P < 0.0001). Our findings present saliva as an applicable alternative for SARS-CoV-2 diagnostics. However, the lower CT values obtained from NPS indicate that NPS may be a slightly more sensitive specimen type. Participants preferred saliva sampling, although delivering an adequate volume of saliva was challenging for some participants. IMPORTANCE The extensive testing of SARS-CoV-2 is vital in controlling the spread of COVID-19. The reference standard for specimen collection is a nasopharyngeal swab (NPS). However, the discomfort of NPS sampling, the risk of nosocomial infections, and global material shortages have accelerated the development of alternative testing methods. Our study demonstrates that patients tolerate saliva sampling better than NPS. Of importance, although the RT-PCR qualitative test results seem to correspond between NPS and saliva, we show significantly lower CT values for NPS, compared to saliva, thus contradicting the suggested superiority of the saliva specimen over NPS in the detection of the Omicron variants of SARS-CoV-2. Future research is still required to enable individual planning for specimen collection and to determine the effects of different SARS-CoV-2 variants on the sensitivity of the saliva matrix.
Collapse
Affiliation(s)
- Jaakko Ahti
- Department of Paediatrics and Adolescent Medicine, University of Turku, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Riikka Österback
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Anniina Keskitalo
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | | | | | - Tytti Vuorinen
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ville Peltola
- Department of Paediatrics and Adolescent Medicine, University of Turku, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Antti J. Hakanen
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Matti Waris
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Miia Laine
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
30
|
Ko Y, Mendoza VM, Mendoza R, Seo Y, Lee J, Jung E. Risk estimation of lifted mask mandates and emerging variants using mathematical model. Heliyon 2023; 9:e16841. [PMID: 37303548 PMCID: PMC10246937 DOI: 10.1016/j.heliyon.2023.e16841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023] Open
Abstract
Background More than half of the population in Korea had a prior COVID-19 infection. In 2022, most nonpharmaceutical interventions, except mask-wearing indoors, had been lifted. And in 2023, the indoor mask mandates were eased. Methods We developed an age-structured compartmental model that distinguishes vaccination history, prior infection, and medical staff from the rest of the population. Contact patterns among hosts were separated based on age and location. We simulated scenarios with the lifting of the mask mandate all at once or sequentially according to the locations. Furthermore, we investigated the impact of a new variant assuming that it has higher transmissibility and risk of breakthrough infection. Results We found that the peak size of administered severe patients may not exceed 1100 when the mask mandate is lifted everywhere, and 800 if the mask mandate only remains in the hospital. If the mask mandate is lifted in a sequence (except hospital), then the peak size of administered severe patients may not exceed 650. Moreover, if the new variant has both higher transmissibility and immune reduction, the effective reproductive number of the new variant is approximately 3 times higher than that of the current variant, and additional interventions may be needed to keep the administered severe patients from exceeding 2,000, which is the critical level we set. Conclusion Our findings showed that the lifting of the mask mandate, except in hospitals, would be more manageable if implemented sequentially. Considering a new variant, we found that depending on the population immunity and transmissibility of the variant, wearing masks and other interventions may be necessary for controlling the disease.
Collapse
Affiliation(s)
- Youngsuk Ko
- Department of Mathematics, Konkuk University, Seoul, South Korea
| | - Victoria May Mendoza
- Department of Mathematics, Konkuk University, Seoul, South Korea
- Institute of Mathematics, University of the Philippines Diliman, Quezon City, Philippines
| | - Renier Mendoza
- Department of Mathematics, Konkuk University, Seoul, South Korea
- Institute of Mathematics, University of the Philippines Diliman, Quezon City, Philippines
| | - Yubin Seo
- Division of Infectious Disease, Department of Internal Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
| | - Jacob Lee
- Division of Infectious Disease, Department of Internal Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
| | - Eunok Jung
- Department of Mathematics, Konkuk University, Seoul, South Korea
| |
Collapse
|
31
|
Barboza VDS, Domingues WB, de Souza TT, Collares TV, Seixas FK, Pacheco BS, Sousa FSS, Oliveira TL, de Lima M, de Pereira CMP, Spilki FR, Giongo JL, Vaucher RDA. Reverse transcription-loop-mediated isothermal amplification (RT-LAMP) assay as a rapid molecular diagnostic tool for COVID-19 in healthcare workers. JOURNAL OF CLINICAL VIROLOGY PLUS 2023; 3:100134. [PMID: 36742065 PMCID: PMC9891106 DOI: 10.1016/j.jcvp.2023.100134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 10/19/2022] [Accepted: 01/06/2023] [Indexed: 01/28/2023] Open
Abstract
In December 2019, the Chinese Center for Disease Control (CDC of China) reported an outbreak of pneumonia in the city of Wuhan (Hubei province, China) that haunted the world, resulting in a global pandemic. This outbreak was caused by a betacoronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Several of these cases have been observed in healthcare professionals working in hospitals and providing care on the pandemic's frontline. In the present study, nasopharyngeal swab samples of healthcare workers were used to assess the performance of the reverse transcription loop-mediated isothermal amplification (RT-LAMP) assay and subsequently compared with the real-time reverse-transcription quantitative PCR (RT-qPCR) method. Thus, in this study, we validated a method for detecting SARS-CoV-2 based on RT-LAMP that can be used to diagnose these workers. The methodology used was based on analyzing the sensitivity, specificity, evaluation of the detection limit, and cross-reaction with other respiratory viruses. The agreement was estimated using a dispersion diagram designed using the Bland-Altman method. A total of 100 clinical specimens of nasopharyngeal swabs were collected from symptomatic and asymptomatic healthcare workers in Pelotas, Brazil, during the SARS-CoV-2 outbreak. RT-LAMP assay, it was possible to detect SARS-CoV-2 in 96.7% of the healthcare professionals tested using the E gene and N gene primers approximately and 100% for the gene of human β-actin. The observed agreement was considered excellent for the primer set of the E and N genes (k = 0.957 and k = 0.896), respectively. The sensitivity of the RT-LAMP assay was positive for the primer set of the E gene, detected to approximately 2 copies per reaction. For the primer set of the N gene, the assay was possible to verify an LoD of approximately 253 copies per reaction. After executing the RT-LAMP assay, no positive reactions were observed for any of the virus respiratory tested. Therefore, we conclude that RT-LAMP is effective for rapid molecular diagnosis during the COVID-19 outbreak period in healthcare professionals.
Collapse
Affiliation(s)
- Victor dos Santos Barboza
- Laboratório de Pesquisa em Bioquímica e Biologia Molecular de Micro-organismos, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - William Borges Domingues
- Laboratório de Genômica Estrutural, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Thobias Toniolo de Souza
- Laboratório de Pesquisa em Bioquímica e Biologia Molecular de Micro-organismos, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Tiago Veiras Collares
- Laboratório de Biotecnologia do Câncer, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Fabiana Kommling Seixas
- Laboratório de Biotecnologia do Câncer, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Bruna Silveira Pacheco
- Laboratório de Biotecnologia do Câncer, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Fernanda Severo Sabedra Sousa
- Laboratório de Biotecnologia do Câncer, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Thaís Larré Oliveira
- Laboratório de Vacinologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Marcelo de Lima
- Laboratório de Virologia e Imunologia, Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | | | - Fernando Rosado Spilki
- Laboratório de Microbiologia Molecular, Universidade FEEVALE, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Janice Luehring Giongo
- Laboratório de Pesquisa em Bioquímica e Biologia Molecular de Micro-organismos, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Rodrigo de Almeida Vaucher
- Laboratório de Pesquisa em Bioquímica e Biologia Molecular de Micro-organismos, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil,Corresponding author
| |
Collapse
|
32
|
Yamamura O, Onishi H, Sakamaki I, Fujita R, Miyashita H, Iwasaki H. Infection rate among close contacts of patients with coronavirus disease in Japan: a descriptive study and literature review. ASIAN BIOMED 2023; 17:115-123. [PMID: 37818161 PMCID: PMC10561678 DOI: 10.2478/abm-2023-0051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023]
Abstract
Background In Japan, on April 20, 2020, the definition of a close contact regarding coronavirus disease (COVID-19) was changed from a long-term contact time to a specified contact time of 15 min and from a contact distance of 2 m to 1 m. Objectives We aimed to determine the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection rate among close contacts of patients with COVID-19 and determine the impact of the infection on transmission among close contacts. Methods The numbers of SARS-CoV-2 tests, SARS-CoV-2-positive cases, and close contacts of patients with COVID-19 were assessed between March 2020 and February 2021 in Fukui Prefecture, Japan. The study period was subdivided into 3 periods. The second and third period contained data with the changed definition of close contact. Results Overall, 32,238 SARS-CoV-2 tests were performed. There were 545 patients with COVID-19 and 1487 close contacts, of whom 267 tested positive. The highest infection rate occurred in period 3. Distance, protective measures, and contact time with COVID-19 patients influenced the increased infection rate. The infection rate showed a rising trend from 11.1% in period 1 to 19.2% and 20.0% in periods 2 and 3, respectively (Cochran-Armitage test; P < 0.004). Multivariate analysis revealed that female sex was an independent risk factor for infection of close contacts (odds ratio: 2.23; 95% confidence interval: 1.700-2.930). Conclusions Female sex is a risk factor for transmission by close contacts. The rate of infection among close contacts may be associated with contact time, contact distance, and protective measures.
Collapse
Affiliation(s)
- Osamu Yamamura
- Department of Community Medicine, Faculty of Medical Sciences, University of Fukui, Fukui910-1193, Japan
| | - Hidenori Onishi
- Department of Community Medicine, Faculty of Medical Sciences, University of Fukui, Fukui910-1193, Japan
| | - Ippei Sakamaki
- Department of Infectious Diseases, Faculty of Medical Sciences, University of Fukui, Fukui910-1193, Japan
| | - Ryousuke Fujita
- Department of Biostatistics, Faculty of Medical Science, University of Fukui, Fukui910-1193, Japan
| | - Hirofumi Miyashita
- Department of Health and Welfare, Fukui Prefecture, Fukui910-8580, Japan
| | - Hiromichi Iwasaki
- Department of Infection Prevention and Control, University of Fukui Hospital, Fukui910-1193, Japan
| |
Collapse
|
33
|
Snipaitiene K, Zablockiene B, Sabaliauskaite R, Zukauskaite K, Matulyte E, Smalinskaite T, Paulauskas M, Zablockis R, Lopeta M, Gagilas J, Puriene A, Jancoriene L, Jarmalaite S. SARS-CoV-2 RT-qPCR Ct values in saliva and nasopharyngeal swab samples for disease severity prediction. J Oral Microbiol 2023; 15:2213106. [PMID: 37213664 PMCID: PMC10193917 DOI: 10.1080/20002297.2023.2213106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/23/2023] Open
Abstract
Background Comparison of clinical value of RT-qPCR-based SARS-CoV-2 tests performed on saliva samples (SSs) and nasopharyngeal swab samples (NPSs) for prediction of the COVID-19 disease severity. Methods Three paired SSs and NPSs collected every 3 days from 100 hospitalised COVID-19 patients during 2020 Jul-2021 Jan were tested by RT-qPCR for the original SARS-CoV-2 virus and compared to 150 healthy controls. Cases were divided into mild+moderate (Cohort I, N = 47) and severe disease (Cohort II, N = 53) cohorts and compared. Results SARS-CoV-2 was detected in 65% (91/140) vs. 53% (82/156) of NPSs and 49% (68/139) vs. 48% (75/157) of SSs collected from Cohort I and II, respectively, resulting in the total respective detection rates of 58% (173/296) vs. 48% (143/296) (P = 0.017). Ct values of SSs were lower than those of NPSs (mean Ct = 28.01 vs. 30.07, P = 0.002). Although Ct values of the first SSs were significantly lower in Cohort I than in Cohort II (P = 0.04), it became negative earlier (mean 11.7 vs. 14.8 days, P = 0.005). Multivariate Cox proportional hazards regression analysis showed that Ct value ≤30 from SSs was the independent predictor for severe COVID-19 (HR = 10.06, 95% CI: 1.84-55.14, P = 0.008). Conclusion Salivary RT-qPCR testing is suitable for SARS-CoV-2 infection control, while simple measurement of Ct values can assist in prediction of COVID-19 severity.
Collapse
Affiliation(s)
- Kristina Snipaitiene
- Institute of Biomedical Sciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Laboratory of Genetic Diagnostic, National Cancer Institute of Lithuania, Vilnius, Lithuania
| | - Birute Zablockiene
- Center of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Clinic of Infectious Diseases and Dermatovenerology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Rasa Sabaliauskaite
- Laboratory of Genetic Diagnostic, National Cancer Institute of Lithuania, Vilnius, Lithuania
| | - Kristina Zukauskaite
- Institute of Biomedical Sciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Laboratory of Genetic Diagnostic, National Cancer Institute of Lithuania, Vilnius, Lithuania
| | - Elzbieta Matulyte
- Center of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Clinic of Infectious Diseases and Dermatovenerology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | | | - Mindaugas Paulauskas
- Center of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Clinic of Infectious Diseases and Dermatovenerology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Rolandas Zablockis
- Clinic of Chest Diseases, Immunology and Allergology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- Centre of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | | | | | - Alina Puriene
- Institute of Odontology, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ligita Jancoriene
- Center of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Clinic of Infectious Diseases and Dermatovenerology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Sonata Jarmalaite
- Institute of Biomedical Sciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Laboratory of Genetic Diagnostic, National Cancer Institute of Lithuania, Vilnius, Lithuania
| |
Collapse
|
34
|
Kim K, Lee B, Park JH, Park JH, Lee KJ, Kwak TJ, Son T, Shin YB, Im H, Kim MG. Rapid PCR kit: lateral flow paper strip with Joule heater for SARS-CoV-2 detection. MATERIALS HORIZONS 2023; 10:1697-1704. [PMID: 36843375 DOI: 10.1039/d2mh01267g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Polymerase chain reaction (PCR)-based diagnostic kits for point-of-care (POC) testing are highly desirable to prevent the spread of infectious diseases. Here, we demonstrate a rapid PCR testing kit that involves integrating a lateral flow paper strip with a nichrome-based thin film heater. The use of a paper membrane as a PCR-solution container results in fast thermocycling without a cooler because the membrane can contain the solution with a high specific surface area where Joule heating is applied. After PCR, amplified products are simultaneously detected at the lateral flow paper strip with the naked eye. Severe acute respiratory syndrome β-coronavirus RNA can be detected within 30 min after PCR solution injection. This work reveals that the paper membrane can act as not only a capillary flow channel but also as a promising platform for fast PCR and detection.
Collapse
Affiliation(s)
- Kihyeun Kim
- Advanced Photonics Research Institute (APRI), Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Bobin Lee
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea.
| | - Jun Hyeok Park
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea.
| | - Ji-Ho Park
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea.
| | - Ki Joong Lee
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Tae Joon Kwak
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Taehwang Son
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Yong-Beom Shin
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- BioNano Health Guard Research Center (H-GUARD), Daejeon 34141, Republic of Korea
| | - Hyungsoon Im
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, 02114, USA
| | - Min-Gon Kim
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea.
| |
Collapse
|
35
|
He M, Xu X, Wang H, Wu Q, Zhang L, Zhou D, Tong Y, Su X, Liu H. Nanozyme-Based Colorimetric SARS-CoV-2 Nucleic Acid Detection by Naked Eye. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2208167. [PMID: 36782092 DOI: 10.1002/smll.202208167] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Indexed: 05/18/2023]
Abstract
Fluorescence-based PCR and other amplification methods have been used for SARS-CoV-2 diagnostics, however, it requires costly fluorescence detectors and probes limiting deploying large-scale screening. Here, a cut-price colorimetric method for SARS-CoV-2 RNA detection by iron manganese silicate nanozyme (IMSN) is established. IMSN catalyzes the oxidation of chromogenic substrates by its peroxidase (POD)-like activity, which is effectively inhibited by pyrophosphate ions (PPi). Due to the large number of PPi generated by amplification processes, SARS-CoV-2 RNA can be detected by a colorimetric readout visible to the naked eye, with the detection limit of 240 copies mL-1 . This conceptually new method has been successfully applied to correctly distinguish positive and negative oropharyngeal swab samples of COVID-19. Colorimetric assay provides a low-cost and instrumental-free solution for nucleic acid detection, which holds great potential for facilitating virus surveillance.
Collapse
Affiliation(s)
- Mengya He
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P.R. China
| | - Xican Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P.R. China
| | - Hongyu Wang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P.R. China
| | - Qingyuan Wu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P.R. China
| | - Linghao Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P.R. China
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, P. R. China
| | - Yigang Tong
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering (BAIC-SM), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xin Su
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P.R. China
| | - Huiyu Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P.R. China
| |
Collapse
|
36
|
Sakai-Tagawa Y, Yamayoshi S, Halfmann PJ, Wilson N, Bobholz M, Vuyk WC, Wei W, Ries H, O'Connor DH, Friedrich TC, Sordillo EM, van Bakel H, Simon V, Kawaoka Y. Sensitivity of rapid antigen tests for Omicron subvariants of SARS-CoV-2. J Med Virol 2023; 95:e28788. [PMID: 37212288 DOI: 10.1002/jmv.28788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/22/2023] [Accepted: 05/02/2023] [Indexed: 05/23/2023]
Abstract
Diagnosis by rapid antigen tests (RATs) is useful for early initiation of antiviral treatment. Because RATs are easy to use, they can be adapted for self-testing. Several kinds of RATs approved for such use by the Japanese regulatory authority are available from drug stores and websites. Most RATs for COVID-19 are based on antibody detection of the SARS-CoV-2 N protein. Since Omicron and its subvariants have accumulated several amino acid substitutions in the N protein, such amino acid changes might affect the sensitivity of RATs. Here, we investigated the sensitivity of seven RATs available in Japan, six of which are approved for public use and one of which is approved for clinical use, for the detection of BA.5, BA.2.75, BF.7, XBB.1, and BQ.1.1, as well as the delta variant (B.1.627.2). All tested RATs detected the delta variant with a detection level between 7500 and 75 000 pfu per test, and all tested RATs showed similar sensitivity to the Omicron variant and its subvariants (BA.5, BA.2.75, BF.7, XBB.1, and BQ.1.1). Human saliva did not reduce the sensitivity of the RATs tested. Espline SARS-CoV-2 N showed the highest sensitivity followed by Inspecter KOWA SARS-CoV-2 and V Trust SARS-CoV-2 Ag. Since the RATs failed to detect low levels of infectious virus, individuals whose specimens contained less infectious virus than the detection limit would be considered negative. Therefore, it is important to note that RATs may miss individuals shedding low levels of infectious virus.
Collapse
Affiliation(s)
- Yuko Sakai-Tagawa
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Seiya Yamayoshi
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Peter J Halfmann
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Nancy Wilson
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Max Bobholz
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - William C Vuyk
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Wanting Wei
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hunter Ries
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David H O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Thomas C Friedrich
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Emilia M Sordillo
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Viviana Simon
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yoshihiro Kawaoka
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Infection and Advanced Research Center, The University of Tokyo Pandemic Preparedness, Tokyo, Japan
| |
Collapse
|
37
|
Fragkou PC, De Angelis G, Menchinelli G, Can F, Garcia F, Morfin-Sherpa F, Dimopoulou D, Dimopoulou K, Zelli S, de Salazar A, Reiter R, Janocha H, Grossi A, Omony J, Skevaki C. Update of ESCMID COVID-19 guidelines: diagnostic testing for SARS-CoV-2. Clin Microbiol Infect 2023:S1198-743X(23)00192-1. [PMID: 37088423 PMCID: PMC10122552 DOI: 10.1016/j.cmi.2023.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/13/2023] [Accepted: 04/16/2023] [Indexed: 04/25/2023]
Abstract
SCOPE Since the onset of coronavirus disease 2019 (COVID-19), several assays have been deployed for the diagnosis of SARS-CoV-2. The European Society of Clinical Microbiology and Infectious Diseases (ESCMID) published the first set of guidelines on SARS-CoV-2 in-vitro diagnosis in February 2022. Since the COVID-19 landscape is rapidly evolving, the relevant ESCMID guidelines panel releases an update of the previously published recommendations on diagnostic testing for SARS-CoV-2. This update aims to delineate the best diagnostic approach for SARS-CoV-2 in different populations based on current evidence. METHODS An ESCMID COVID-19 guidelines task force was established by the ESCMID Executive Committee. A small group was established, half appointed by the chair, and the remaining selected with an open call. The panel met virtually once a week. For all decisions, a simple majority vote was used. A list of clinical questions using the PICO (population, intervention, comparison, and outcome) format was developed at the beginning of the process. For each PICO, two panel members performed a literature search focusing on systematic reviews with a third panellist involved in case of inconsistent results. The panel reassessed the PICOs previously defined as priority in the first set of guidelines and decided to address 49 PICO questions, as 6 of them were discarded as outdated/non-clinically relevant. The "Grading of Recommendations Assessment, Development and Evaluation(GRADE)-adoption, adaptation, and de novo development of recommendations (ADOLOPMENT)" evidence-to-decision framework was utilized to produce the guidelines. QUESTIONS ADDRESSED BY THE GUIDELINE AND RECOMMENDATIONS After literature search, we updated 16 PICO questions; these PICOs address the use of antigen-based assays among symptomatic and asymptomatic patients with different ages, COVID-19 severity status or risk for severe COVID-19, time since onset of symptoms/contact with an infectious case, and finally, types of biomaterials used.
Collapse
Affiliation(s)
- Paraskevi C Fragkou
- First Department of Critical Care Medicine & Pulmonary Services, Evangelismos General Hospital, National and Kapodistrian University of Athens, Athens, Greece; European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Respiratory Viruses (ESGREV)
| | - Giulia De Angelis
- European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Respiratory Viruses (ESGREV); Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS - 00168, Rome, Italy
| | - Giulia Menchinelli
- European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Respiratory Viruses (ESGREV); Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS - 00168, Rome, Italy; Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Fusun Can
- European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Respiratory Viruses (ESGREV); Department of Medical Microbiology, Koc University School of Medicine, Istanbul, Turkey; Koc University IsBank Research Centre for Infectious Diseases (KUISCID), Istanbul, Turkey
| | - Federico Garcia
- European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Respiratory Viruses (ESGREV); Servicio de Microbiología Clínica. Hospital Universitario Clínico San Cecilio. Instituto de Investigación Biosanitaria, Ibs.GRANADA, Granada, Spain; Centro de Investigación Biomédicaen Red Enfermedades Infecciosas (CIBERINFEC), ISCIII, Madrid, Spain
| | - Florence Morfin-Sherpa
- European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Respiratory Viruses (ESGREV); Laboratory of Virology, Institut des Agents Infectieux, National Reference Centre for respiratory viruses, Hospices Civils de Lyon, Université Claude Bernard Lyon1, Lyon, France
| | - Dimitra Dimopoulou
- European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Respiratory Viruses (ESGREV); Second Department of Paediatrics, "P. and A. Kyriakou" Children's Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Silvia Zelli
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS - 00168, Rome, Italy
| | - Adolfo de Salazar
- Servicio de Microbiología Clínica. Hospital Universitario Clínico San Cecilio. Instituto de Investigación Biosanitaria, Ibs.GRANADA, Granada, Spain; Centro de Investigación Biomédicaen Red Enfermedades Infecciosas (CIBERINFEC), ISCIII, Madrid, Spain
| | - Rieke Reiter
- Institute of Laboratory Medicine, Universities of Giessen and Marburg Lung Centre (UGMLC), Philipps University Marburg, German Centre for Lung Research (DZL), Marburg, Germany
| | - Hannah Janocha
- Institute of Laboratory Medicine, Universities of Giessen and Marburg Lung Centre (UGMLC), Philipps University Marburg, German Centre for Lung Research (DZL), Marburg, Germany
| | | | - Jimmy Omony
- Institute for Asthma and Allergy Prevention (IAP), Helmholtz Zentrum Munich, German Research Centre for Environmental Health (GmbH), Munich, Germany
| | - Chrysanthi Skevaki
- European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Respiratory Viruses (ESGREV); Institute of Laboratory Medicine, Universities of Giessen and Marburg Lung Centre (UGMLC), Philipps University Marburg, German Centre for Lung Research (DZL), Marburg, Germany.
| |
Collapse
|
38
|
Nahar K, Begum MN, Tony SR, Jubair M, Hossain MA, Karim Y, Faisal AA, Hossain ME, Rahman MZ, Rahman M. Nasal swab as an alternative specimen for the detection of severe acute respiratory syndrome coronavirus 2. Health Sci Rep 2023; 6:e1213. [PMID: 37077182 PMCID: PMC10106929 DOI: 10.1002/hsr2.1213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/06/2023] [Accepted: 04/06/2023] [Indexed: 04/21/2023] Open
Abstract
Background and Aims The coronavirus disease 2019 (COVID-19) has brought serious threats to public health worldwide. Nasopharyngeal, nasal swabs, and saliva specimens are used to detect severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, limited data are available on the performance of less invasive nasal swab for testing COVID-19. This study aimed to compare the diagnostic performance of nasal swabs with nasopharyngeal swabs using real-time reverse transcription polymerase chain reaction (RT-PCR) considering viral load, onset of symptoms, and disease severity. Methods A total of 449 suspected COVIDCOVID-19 individuals were recruited. Both nasopharyngeal and nasal swabs were collected from the same individual. Viral RNA was extracted and tested by real-time RT-PCR. Metadata were collected using structured questionnaire and analyzed by SPSS and MedCalc software. Results The overall sensitivity of the nasopharyngeal swab was 96.6%, and the nasal swab was 83.4%. The sensitivity of nasal swabs was more than 97.7% for low and moderate C t values. Moreover, the performance of nasal swab was very high (>87%) for hospitalized patients and at the later stage >7 days of onset of symptoms. Conclusion Less invasive nasal swab sampling with adequate sensitivity can be used as an alternative to nasopharyngeal swabs for the detection of SARS-CoV-2 by real-time RT-PCR.
Collapse
Affiliation(s)
- Kamrun Nahar
- Virology LaboratoryInfectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, BangladeshMohakhaliDhaka1212Bangladesh
| | - Mst. Noorjahan Begum
- Virology LaboratoryInfectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, BangladeshMohakhaliDhaka1212Bangladesh
| | - Selim R. Tony
- Virology LaboratoryInfectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, BangladeshMohakhaliDhaka1212Bangladesh
| | - Mohammad Jubair
- Virology LaboratoryInfectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, BangladeshMohakhaliDhaka1212Bangladesh
| | - Md. Abir Hossain
- Virology LaboratoryInfectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, BangladeshMohakhaliDhaka1212Bangladesh
| | - Yeasir Karim
- Virology LaboratoryInfectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, BangladeshMohakhaliDhaka1212Bangladesh
| | - Abdullah Al. Faisal
- Virology LaboratoryInfectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, BangladeshMohakhaliDhaka1212Bangladesh
| | - Mohammad Enayet Hossain
- Virology LaboratoryInfectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, BangladeshMohakhaliDhaka1212Bangladesh
| | - Mohammed Ziaur Rahman
- Virology LaboratoryInfectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, BangladeshMohakhaliDhaka1212Bangladesh
| | - Mustafizur Rahman
- Virology LaboratoryInfectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, BangladeshMohakhaliDhaka1212Bangladesh
| |
Collapse
|
39
|
Gao P, Duan W, Shi H, Wang Q. Silencing circPalm2 inhibits sepsis-induced acute lung injury by sponging miR-376b-3p and targeting MAP3K1. Toxicol Res 2023; 39:275-294. [PMID: 37008689 PMCID: PMC10050541 DOI: 10.1007/s43188-022-00169-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
The apoptosis and inflammation of pulmonary epithelial cells are important pathogenic factors of sepsis-induced acute lung injury (ALI). Upregulation of circPalm2 (circ_0001212) expression levels has been previously detected in the lung tissue of ALI rats. Herein, the biological significance and detailed mechanism of circPalm2 in ALI pathogenesis were investigated. In vivo models of sepsis-induced ALI were established by treating C57BL/6 mice with cecal ligation and puncture (CLP) surgery. Murine pulmonary epithelial cells (MLE-12 cells) were stimulated with lipopolysaccharide (LPS) to establish in vitro septic ALI models. MLE-12 cell viability and apoptosis were evaluated by CCK-8 assay and flow cytometry analysis, respectively. The pathological alterations of the lung tissue were analysed based on hematoxylin-eosin (H&E) staining. Cell apoptosis in the lung tissue samples was examined by TUNEL staining assay. LPS administration suppressed the viability and accelerated the inflammation and apoptotic behaviours of MLE-12 cells. CircPalm2 displayed high expression in LPS-stimulated MLE-12 cells and possessed circular characteristics. The silencing of circPalm2 impeded apoptosis and inflammation in LPS-stimulated MLE-12 cells. Mechanistically, circPalm2 bound with miR-376b-3p, which targeted MAP3K1. In rescue assays, MAP3K1 enhancement reversed the repressive effects of circPalm2 depletion on LPS-triggered inflammatory injury and MLE-12 cell apoptosis. Furthermore, the lung tissue collected from CLP model mice displayed low miR-376b-3p expression and high levels of circPalm2 and MAP3K1. CircPalm2 positively regulated MAP3K1 expression by downregulating miR-376b-3p in murine lung tissues. Importantly, circPalm2 knockdown attenuated CLP-induced inflammation, apoptosis, and pathological alterations in lung tissues collected from mice. Silenced circPalm2 inhibits LPS-induced pulmonary epithelial cell dysfunction and mitigates abnormalities in lung tissues collected from CLP-stimulated mice via the miR-376b-3p/MAP3K1 axis in septic ALI. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-022-00169-7.
Collapse
Affiliation(s)
- Pengfei Gao
- Shanghai East Clinical Medical College, Nanjing Medical University, No. 150, Jimo Road, Pudong New Area, Shanghai, 200120 China
- Department of Anesthesiology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, Jiangsu 223300 China
| | - Wenying Duan
- Shanghai East Clinical Medical College, Nanjing Medical University, No. 150, Jimo Road, Pudong New Area, Shanghai, 200120 China
| | - Huiyan Shi
- Jinzhou Medical University, Jinzhou, Liaoning 121001 China
| | - Qingxiu Wang
- Shanghai East Clinical Medical College, Nanjing Medical University, No. 150, Jimo Road, Pudong New Area, Shanghai, 200120 China
- Shanghai East Hopital, Tongji University School of Medicine, Shanghai, 200120 China
| |
Collapse
|
40
|
Szobi A, Buranovská K, Vojtaššáková N, Lovíšek D, Özbaşak HÖ, Szeibeczederová S, Kapustian L, Hudáčová Z, Kováčová V, Drobná D, Putaj P, Bírová S, Čirková I, Čarnecký M, Kilián P, Jurkáček P, Čabanová V, Boršová K, Sláviková M, Vaňová V, Klempa B, Čekan P, Paul ED. Vivid COVID-19 LAMP is an ultrasensitive, quadruplexed test using LNA-modified primers and a zinc ion and 5-Br-PAPS colorimetric detection system. Commun Biol 2023; 6:233. [PMID: 36864129 PMCID: PMC9979146 DOI: 10.1038/s42003-023-04612-9] [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: 01/20/2023] [Accepted: 02/20/2023] [Indexed: 03/04/2023] Open
Abstract
Sensitive and rapid point-of-care assays have been crucial in the global response to SARS-CoV-2. Loop-mediated isothermal amplification (LAMP) has emerged as an important diagnostic tool given its simplicity and minimal equipment requirements, although limitations exist regarding sensitivity and the methods used to detect reaction products. We describe the development of Vivid COVID-19 LAMP, which leverages a metallochromic detection system utilizing zinc ions and a zinc sensor, 5-Br-PAPS, to circumvent the limitations of classic detection systems dependent on pH indicators or magnesium chelators. We make important strides in improving RT-LAMP sensitivity by establishing principles for using LNA-modified LAMP primers, multiplexing, and conducting extensive optimizations of reaction parameters. To enable point-of-care testing, we introduce a rapid sample inactivation procedure without RNA extraction that is compatible with self-collected, non-invasive gargle samples. Our quadruplexed assay (targeting E, N, ORF1a, and RdRP) reliably detects 1 RNA copy/µl of sample (=8 copies/reaction) from extracted RNA and 2 RNA copies/µl of sample (=16 copies/reaction) directly from gargle samples, making it one of the most sensitive RT-LAMP tests and even comparable to RT-qPCR. Additionally, we demonstrate a self-contained, mobile version of our assay in a variety of high-throughput field testing scenarios on nearly 9,000 crude gargle samples. Vivid COVID-19 LAMP can be an important asset for the endemic phase of COVID-19 as well as preparing for future pandemics.
Collapse
Affiliation(s)
- Adrián Szobi
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Katarína Buranovská
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Nina Vojtaššáková
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Daniel Lovíšek
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Halil Önder Özbaşak
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Sandra Szeibeczederová
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Liudmyla Kapustian
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Zuzana Hudáčová
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
- Stanford University, 730 Escondido Rd., Stanford, CA, 94305, USA
| | - Viera Kováčová
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
- University of Cologne, Institute for Biological Physics, Zülpicher Str. 77, 50937, Köln, Germany
| | - Diana Drobná
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Piotr Putaj
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Stanislava Bírová
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Ivana Čirková
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Martin Čarnecký
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Peter Kilián
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA
| | - Peter Jurkáček
- AstonITM s.r.o., Račianska 153, 831 54, Bratislava, Slovakia
| | - Viktória Čabanová
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Virology, Dúbravská cesta 9, 845 05, Bratislava, Slovakia
| | - Kristína Boršová
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Virology, Dúbravská cesta 9, 845 05, Bratislava, Slovakia
| | - Monika Sláviková
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Virology, Dúbravská cesta 9, 845 05, Bratislava, Slovakia
| | - Veronika Vaňová
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Virology, Dúbravská cesta 9, 845 05, Bratislava, Slovakia
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University, Ilkovičova 6, 842 15, Bratislava, Slovakia
| | - Boris Klempa
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Virology, Dúbravská cesta 9, 845 05, Bratislava, Slovakia
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University, Ilkovičova 6, 842 15, Bratislava, Slovakia
| | - Pavol Čekan
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia.
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA.
| | - Evan D Paul
- MultiplexDX, s.r.o., Comenius University Science Park, Ilkovičova 8, 841 04, Bratislava, Slovakia.
- MultiplexDX, Inc., One Research Court, Suite 450, Rockville, MD, 20850, USA.
| |
Collapse
|
41
|
Cai Y, Liu M, Wu Z, Tian C, Qiu S, Li Z, Xu F, Li W, Zheng Y, Xu A, Xie L, Tan X. Diagnostic accuracy of autoverification and guidance system for COVID-19 RT-PCR results. EPMA J 2023; 14:119-129. [PMID: 36540610 PMCID: PMC9755791 DOI: 10.1007/s13167-022-00310-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/05/2022] [Indexed: 12/21/2022]
Abstract
BACKGROUND To date, most countries worldwide have declared that the pandemic of COVID-19 is over, while the WHO has not officially ended the COVID-19 pandemic, and China still insists on the personalized dynamic COVID-free policy. Large-scale nucleic acid testing in Chinese communities and the manual interpretation for SARS-CoV-2 nucleic acid detection results pose a huge challenge for labour, quality and turnaround time (TAT) requirements. To solve this specific issue while increase the efficiency and accuracy of interpretation, we created an autoverification and guidance system (AGS) that can automatically interpret and report the COVID-19 reverse transcriptase-polymerase chain reaction (RT-PCR) results relaying on computer-based autoverification procedure and then validated its performance in real-world environments. This would be conductive to transmission risk prediction, COVID-19 prevention and control and timely medical treatment for positive patients in the context of the predictive, preventive and personalized medicine (PPPM). METHODS A diagnostic accuracy test was conducted with 380,693 participants from two COVID-19 test sites in China, the Hong Kong Hybribio Medical Laboratory (n = 266,035) and the mobile medical shelter at a Shanghai airport (n = 114,658). These participants underwent SARS-CoV-2 RT-PCR from March 28 to April 10, 2022. All RT-PCR results were interpreted by laboratorians and by using AGS simultaneously. Considering the manual interpretation as gold standard, the sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV) and accuracy were applied to evaluate the diagnostic value of the AGS on the interpretation of RT-PCR results. RESULTS Among the 266,035 samples in Hong Kong, there were 16,356 (6.15%) positive, 231,073 (86.86%) negative, 18,606 (6.99%) indefinite, 231,073 (86.86%, negative) no retest required and 34,962 (13.14%, positive and indefinite) retest required; the 114,658 samples in Shanghai consisted of 76 (0.07%) positive, 109,956 (95.90%) negative, 4626 (4.03%) indefinite, 109,956 (95.90%, negative) no retest required and 4702 (4.10%, positive and indefinite) retest required. Compared to the fashioned manual interpretation, the AGS is a procedure of high accuracy [99.96% (95%CI, 99.95-99.97%) in Hong Kong and 100% (95%CI, 100-100%) in Shanghai] with perfect sensitivity [99.98% (95%CI, 99.97-99.98%) in Hong Kong and 100% (95%CI, 100-100%) in Shanghai], specificity [99.87% (95%CI, 99.82-99.90%) in Hong Kong and 100% (95%CI, 99.92-100%) in Shanghai], PPV [99.98% (95%CI, 99.97-99.99%) in Hong Kong and 100% (95%CI, 99.99-100%) in Shanghai] and NPV [99.85% (95%CI, 99.80-99.88%) in Hong Kong and 100% (95%CI, 99.90-100%) in Shanghai]. The need for manual interpretation of total samples was dramatically reduced from 100% to 13.1% and the interpretation time fell from 53 h to 26 min in Hong Kong; while the manual interpretation of total samples was decreased from 100% to 4.1% and the interpretation time dropped from 20 h to 16 min at Shanghai. CONCLUSIONS The AGS is a procedure of high accuracy and significantly relieves both labour and time from the challenge of large-scale screening of SARS-CoV-2 using RT-PCR. It should be recommended as a powerful screening, diagnostic and predictive system for SARS-CoV-2 to contribute timely the ending of the COVID-19 pandemic following the concept of PPPM.
Collapse
Affiliation(s)
- Yingmu Cai
- Joint Laboratory of Shantou University Medical College and Guangdong Hybribio Biotech Ltd, Shantou University Medical College, Shantou, 515041 Guangdong China
- Hybribio Medical Laboratory Group Ltd, Chaozhou, 521000 Guangdong China
- Clinical Research Centre, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Mengyu Liu
- Joint Laboratory of Shantou University Medical College and Guangdong Hybribio Biotech Ltd, Shantou University Medical College, Shantou, 515041 Guangdong China
- Clinical Research Centre, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Zhiyuan Wu
- Beijing Municipal Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, 100069 China
- Centre for Precision Health, Edith Cowan University, Perth, WA 6027 Australia
| | - Cuihong Tian
- Clinical Research Centre, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
- Centre for Precision Health, Edith Cowan University, Perth, WA 6027 Australia
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Song Qiu
- Hybribio Medical Laboratory Group Ltd, Chaozhou, 521000 Guangdong China
| | - Zhen Li
- Human Papillomavirus Molecular Diagnostic Engineering Technology Research Centre, Chaozhou, 521000 Guangdong China
| | - Feng Xu
- Human Papillomavirus Molecular Diagnostic Engineering Technology Research Centre, Chaozhou, 521000 Guangdong China
| | - Wei Li
- Joint Laboratory of Shantou University Medical College and Guangdong Hybribio Biotech Ltd, Shantou University Medical College, Shantou, 515041 Guangdong China
- Clinical Research Centre, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Yan Zheng
- Department of Research and Development, Guangdong Research Institute of Genetic Diagnostic and Engineering Technologies for Thalassemia, Chaozhou, 521011 Guangdong China
| | - Aijuan Xu
- Human Papillomavirus Molecular Diagnostic Engineering Technology Research Centre, Chaozhou, 521000 Guangdong China
| | - Longxu Xie
- Hybribio Medical Laboratory Group Ltd, Chaozhou, 521000 Guangdong China
- Human Papillomavirus Molecular Diagnostic Engineering Technology Research Centre, Chaozhou, 521000 Guangdong China
| | - Xuerui Tan
- Clinical Research Centre, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| |
Collapse
|
42
|
Chirico F, Yıldırım M, Dzieciatkowski T, Dabrowski M, Malysz M, Madziala M, Jaguszewski MJ, Bielski K, Nucera G, Filipiak KJ, Szarpak L. Efficiency rating of SG Diagnostics COVID-19 antigen rapid test kit. Future Virol 2023:10.2217/fvl-2021-0210. [PMID: 37091964 PMCID: PMC10115194 DOI: 10.2217/fvl-2021-0210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/16/2023] [Indexed: 04/25/2023]
Abstract
Aim: Rapid detection is crucial in complementing vaccination to reduce transmission of SARS-CoV-2. Materials & methods: Nasopharyngeal swabs (n = 213) and oropharyngeal swabs (n = 98) were tested. with the antigen rapid test kit. Results: Overall sensitivity (97.96%), specificity (100.00%) and coincidence rate (98.71%) were high, which translated into a positive predictive value of 100.00% and a negative predictive value of 96.64%. Conclusion: Antigen rapid tests have a great potential for screening in different settings to deliver results with high sensitivity and specificity.
Collapse
Affiliation(s)
- Francesco Chirico
- Post-graduate School of Occupational Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Health Service Department, Italian State Police, Ministry of the Interior, Milan, Italy
- Author for correspondence:
| | - Murat Yıldırım
- Department of Psychology, Faculty of Science & Letters, Agri Ibrahim Cecen University, Ağrı, Turkey
| | - Tomasz Dzieciatkowski
- Chair & Department of Medical Microbiology, Medical University of Warsaw, Warsaw, Poland
| | - Marek Dabrowski
- Research Unit, Polish Society of Disaster Medicine, Warsaw, Poland
- Chair & Department of Medical Education, Poznan University of Medical Sciences, Poznan, Poland
| | - Marek Malysz
- Research Unit, Polish Society of Disaster Medicine, Warsaw, Poland
- Institute of Outcomes Research, Polonia University, Czestochowa, Poland
| | - Marcin Madziala
- Research Unit, Polish Society of Disaster Medicine, Warsaw, Poland
| | | | - Karol Bielski
- Research Unit, Polish Society of Disaster Medicine, Warsaw, Poland
- Institute of Outcomes Research, Polonia University, Czestochowa, Poland
- Emergency Medical Service, Warsaw, Poland
| | - Gabriella Nucera
- Department of Emergency, Fatebenefratelli Hospital, ASST Fatebenefratelli & Sacco, Milan, Italy
| | - Krzysztof J Filipiak
- Institute of Outcomes Research, Maria Sklodowska-Curie Medical Academy, Warsaw, Poland
| | - Lukasz Szarpak
- Research Unit, Polish Society of Disaster Medicine, Warsaw, Poland
- Institute of Outcomes Research, Maria Sklodowska-Curie Medical Academy, Warsaw, Poland
- Research Unit, Maria Sklodowska-Curie Bialystok Oncology Center, Bialystok, Poland
| |
Collapse
|
43
|
Kodana M, Orihara Y, Tezuka M, Takahashi R, Noguchi S, Matsuzaki N, Takada T, Kobari N, Ogane K, Kawamura R, Kawamura T, Takeuchi S, Kamiyama Y, Shiomi R, Aoyagi R, Saito M, Kusano T, Nakaya N, Kaneko S, Morita H, Uchida Y, Yazawa H, Sekiya R, Katayama K, Mikami S, Sato T, Tarumoto N, Kobayashi T, Nakamoto H, Maeda T. Evaluation of an immunochromatography-based rapid antigen test, Inspecter Kowa® SARS-CoV-2, using saliva specimens for the detection of SARS-CoV-2. J Infect Chemother 2023; 29:586-591. [PMID: 36849098 PMCID: PMC9968476 DOI: 10.1016/j.jiac.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 01/17/2023] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
BACKGROUND In the context of the coronavirus disease 2019 (COVID-19) pandemic, a rapid and reliable point-of-care test is an essential tool for controlling the spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In particular, an immunochromatography test (ICT) that uses saliva specimens for rapid antigen detection not only reduces the risk of secondary infections but also reduces the burden on medical personnel. METHODS The newly developed salivary antigen test kit "Inspecter Kowa® SARS-CoV-2" is an ICT to which saliva specimens can be directly applied. We evaluated its usefulness in comparison with reverse transcription quantitative PCR (RT-qPCR) and the Espline® SARS-CoV-2 Kit for the detection of SARS-CoV-2 using nasopharyngeal swab specimens. In this study, 140 patients with suspected symptomatic COVID-19 who visited our hospital were enrolled, and nasopharyngeal swab and saliva specimens were collected after they consented to participate in the study. RESULTS Inspector Kowa SARS-CoV-2 was positive in 45 of 61 (73.8%) saliva that were positive by RT-qPCR and the Espline® SARS-CoV-2 Kit was also positive in 56 of 60 (93.3%) Np swabs that were positive by RT-qPCR. Good antigen detection was achieved by ICT with saliva and nasopharyngeal swab specimens when viral load was ≥105 copies/mL, whereas detection sensitivity was low when viral load was <105 copies/mL, especially in saliva specimens. CONCLUSION This ICT for the detection of SARS-CoV-2 salivary antigen is an attractive tool that does not require specialized equipment and allows patients to perform the entire process from sample collection to self-diagnose and to reduce the burden on medical care during a pandemic.
Collapse
Affiliation(s)
- Masahiro Kodana
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan.
| | - Yuta Orihara
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Mariko Tezuka
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Rina Takahashi
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Sakiko Noguchi
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Nanako Matsuzaki
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Tomohito Takada
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Naomi Kobari
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Kana Ogane
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Rieko Kawamura
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Toru Kawamura
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Shinichi Takeuchi
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Yuki Kamiyama
- Department of General Internal Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Rie Shiomi
- Department of General Internal Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Ryutaro Aoyagi
- Department of General Internal Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Masaya Saito
- Department of General Internal Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Takeru Kusano
- Department of General Internal Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Nobuaki Nakaya
- Department of General Internal Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Satoru Kaneko
- Department of Nephrology, Saitama Medical University Hospital, Saitama, Japan
| | - Hideo Morita
- Department of Endocrinology and Diabetes, Saitama Medical University Hospital, Saitama, Japan
| | - Yoshihito Uchida
- Department of Gastroenterology and Hepatology, Saitama Medical University Hospital, Saitama, Japan
| | - Hiroaki Yazawa
- Department of Rheumatology and Applied Immunology, Saitama Medical University Hospital, Saitama, Japan
| | - Ryu Sekiya
- Department of Respiratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Kazuki Katayama
- Department of Respiratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Shingo Mikami
- Department of Neurology, Saitama Medical University Hospital, Saitama, Japan
| | - Tomoya Sato
- Department of Plastic, Reconstructive and Aesthetic Surgery, Saitama Medical University Hospital, Saitama, Japan
| | - Norihito Tarumoto
- Department of Infectious Disease and Infection Control, Saitama Medical University Hospital, Saitama, Japan
| | - Takehito Kobayashi
- Department of General Internal Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Hidetomo Nakamoto
- Department of General Internal Medicine, Saitama Medical University Hospital, Saitama, Japan
| | - Takuya Maeda
- Department of Clinical Laboratory Medicine, Saitama Medical University Hospital, Saitama, Japan
| |
Collapse
|
44
|
Matsumura Y, Yamazaki W, Noguchi T, Yamamoto M, Nagao M. Analytical and clinical performances of seven direct detection assays for SARS-CoV-2. JOURNAL OF CLINICAL VIROLOGY PLUS 2023; 3:100138. [PMID: 36683610 PMCID: PMC9837381 DOI: 10.1016/j.jcvp.2023.100138] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/28/2022] [Accepted: 01/12/2023] [Indexed: 01/14/2023] Open
Abstract
Background Direct detection tests for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that bypass complicated nucleic acid/antigen purification steps are promising tools for the rapid diagnosis of coronavirus disease 2019 (COVID-19). Methods To determine the analytical and clinical diagnostic performances of the direct detection assays, we compared 6 direct molecular detection assays, including two loop-mediated isothermal amplification (LAMP) assays and one lateral flow antigen assay, against the reference extraction-based RT-PCR assay using 183 respiratory samples (87 nasopharyngeal swabs, 51 saliva samples, and 45 sputum samples). Results Analytical sensitivity analysis showed that the direct RT-PCR assay of Toyobo exhibited the lowest LOD of 1,000 copies/mL. Compared with the 80 positive and 103 negative samples based on the reference assay, the Toyobo assay had the highest positive percent agreement (PPA) of 96.3%, followed by the two direct RT-PCR assays of Takara and Shimadzu and one LAMP assay of Eiken (86.3-87.5%). The Fujirebio antigen assay had the lowest PPA of 44.7% among the assays tested. The negative percent agreement of these direct detection assays was 100%, except for the Eiken assay (96.3%). Conclusions Large differences in PPA existed among the direct detection tests. Laboratories need to take these characteristics into consideration before implementing these assays.
Collapse
Affiliation(s)
- Yasufumi Matsumura
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-kawaharacho, Sakyo-ku, Kyoto, 6068507, Japan,Corresponding author at: Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-kawaharacho, Sakyo-ku, Kyoto 6068507, Japan
| | - Wataru Yamazaki
- Center for Southeast Asian Studies, Kyoto University, 46 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 6068501, Japan
| | - Taro Noguchi
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-kawaharacho, Sakyo-ku, Kyoto, 6068507, Japan
| | - Masaki Yamamoto
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-kawaharacho, Sakyo-ku, Kyoto, 6068507, Japan
| | - Miki Nagao
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-kawaharacho, Sakyo-ku, Kyoto, 6068507, Japan
| |
Collapse
|
45
|
Li S, Guo W, Xiao M, Chen Y, Luo X, Xu W, Zhou J, Wang J. Rapid and Sensitive Diagnosis of COVID-19 Using an Electricity-Free Self-Testing System. BIOSENSORS 2023; 13:180. [PMID: 36831946 PMCID: PMC9953845 DOI: 10.3390/bios13020180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 06/18/2023]
Abstract
Rapid and sensitive detection of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is essential for early diagnosis and effective treatment. Nucleic acid testing has been considered the gold standard method for the diagnosis of COVID-19 for its high sensitivity and specificity. However, the polymerase chain reaction (PCR)-based method in the central lab requires expensive equipment and well-trained personnel, which makes it difficult to be used in resource-limited settings. It highlights the need for a sensitive and simple assay that allows potential patients to detect SARS-CoV-2 by themselves. Here, we developed an electricity-free self-testing system based on reverse transcription loop-mediated isothermal amplification (RT-LAMP) that allows for rapid and accurate detection of SARS-CoV-2. Our system employs a heating bag as the heat source, and a 3D-printed box filled with phase change material (PCM) that successfully regulates the temperature for the RT-LAMP. The colorimetric method could be completed in 40 min and the results could be read out by the naked eye. A ratiometric measurement for exact readout was also incorporated to improve the detection accuracy of the system. This self-testing system is a promising tool for point-of-care testing (POCT) that enables rapid and sensitive diagnosis of SARS-CoV-2 in the real world and will improve the current COVID-19 screening efforts for control and mitigation of the pandemic.
Collapse
Affiliation(s)
- Sheng Li
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Wenlong Guo
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Minmin Xiao
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Yulin Chen
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Xinyi Luo
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Wenfei Xu
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China
| | - Jianhua Zhou
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Jiasi Wang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
46
|
Wey L, Masetto T, Spaeth A, Brehm J, Kochem C, Reinhart M, Müller H, Kempin U, Lorenz F, Peter C, Grimmler M. Bioinformatical Design and Performance Evaluation of a Nucleocapsid- and an RBD-Based Particle Enhanced Turbidimetric Immunoassay (PETIA) to Quantify the Wild Type and Variants of Concern-Derived Immunoreactivity of SARS-CoV-2. Biomedicines 2023; 11:160. [PMID: 36672668 PMCID: PMC9855841 DOI: 10.3390/biomedicines11010160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/29/2022] [Accepted: 12/31/2022] [Indexed: 01/11/2023] Open
Abstract
Since SARS-CoV-2 emerged in December 2019 in Wuhan, the resulting pandemic has paralyzed the economic and cultural life of the world. Variants of concern (VOC) strongly increase pressure on public health systems. Rapid, easy-to-use, and cost-effective assays are essential to manage the pandemic. Here we present a bioinformatical approach for the fast and efficient design of two innovative serological Particle Enhanced Turbidimetric Immunoassays (PETIA) to quantify the SARS-CoV-2 immunoresponse. To confirm bioinformatical assumptions, an S-RBD- and a Nucleocapsid-based PETIA were produced. Sensitivity and specificity were compared for 95 patient samples using a BioMajesty™ fully automated analyzer. The S-RBD-based PETIA showed necessary specificity (98%) over the N protein-based PETIA (21%). Further, the reactivity and cross-reactivity of the RBD-based PETIA towards variant-derived antibodies of SARS-CoV-2 were assessed by a quenching inhibition test. The inhibition kinetics of the S-RBD variants Alpha, Beta, Delta, Gamma, Kappa, and Omicron were evaluated. In summary, we showed that specific and robust PETIA immunoassays can be rapidly designed and developed. The quantification of the SARS-CoV-2-related immunoresponse of variants (Alpha to Kappa) is possible using specific RBD assays. In contrast, Omicron revealed lower cross-reactivity (approx. 50%). To ensure the quantification of the Omicron variant, modified immunoassays appear to be necessary.
Collapse
Affiliation(s)
- Leoni Wey
- DiaSys Diagnostic Systems GmbH, Alte Str. 9, 65558 Holzheim, Germany
- Hochschule Fresenius Gem. Trägergesellschaft mbH, University of Applied Sciences, Limburger Str. 2, 65510 Idstein, Germany
| | - Thomas Masetto
- DiaSys Diagnostic Systems GmbH, Alte Str. 9, 65558 Holzheim, Germany
- Institut für Molekulare Medizin I, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Alexander Spaeth
- MVZ Medizinische Labore Dessau Kassel GmbH, Bauhüttenstr. 6, 06847 Dessau-Roßlau, Germany
| | - Jessica Brehm
- MVZ Medizinische Labore Dessau Kassel GmbH, Bauhüttenstr. 6, 06847 Dessau-Roßlau, Germany
| | - Christian Kochem
- DiaSys Diagnostic Systems GmbH, Alte Str. 9, 65558 Holzheim, Germany
| | | | - Holger Müller
- DiaSys Diagnostic Systems GmbH, Alte Str. 9, 65558 Holzheim, Germany
| | - Uwe Kempin
- pes Medizinische Diagnosesysteme GmbH, Hauptstr. 103, 04416 Markkleeberg, Germany
| | - Franziska Lorenz
- MVZ Medizinische Labore Dessau Kassel GmbH, Bauhüttenstr. 6, 06847 Dessau-Roßlau, Germany
| | - Christoph Peter
- Institut für Molekulare Medizin I, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Matthias Grimmler
- DiaSys Diagnostic Systems GmbH, Alte Str. 9, 65558 Holzheim, Germany
- Hochschule Fresenius Gem. Trägergesellschaft mbH, University of Applied Sciences, Limburger Str. 2, 65510 Idstein, Germany
| |
Collapse
|
47
|
Pittman TW, Decsi DB, Punyadeera C, Henry CS. Saliva-based microfluidic point-of-care diagnostic. Theranostics 2023; 13:1091-1108. [PMID: 36793864 PMCID: PMC9925318 DOI: 10.7150/thno.78872] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
There has been a long-standing interest in point-of-care (POC) diagnostics as a tool to improve patient care because it can provide rapid, actionable results near the patient. Some of the successful examples of POC testing include lateral flow assays, urine dipsticks, and glucometers. Unfortunately, POC analysis is somewhat limited by the ability to manufacture simple devices to selectively measure disease specific biomarkers and the need for invasive biological sampling. Next generation POCs are being developed that make use of microfluidic devices to detect biomarkers in biological fluids in a non-invasive manner, addressing the above-mentioned limitations. Microfluidic devices are desirable because they can provide the ability to perform additional sample processing steps not available in existing commercial diagnostics. As a result, they can provide more sensitive and selective analysis. While most POC methods make use of blood or urine as a sample matrix, there has been a growing push to use saliva as a diagnostic medium. Saliva represents an ideal non-invasive biofluid for detecting biomarkers because it is readily available in large quantities and analyte levels reflect those in blood. However, using saliva in microfluidic devices for POC diagnostics is a relatively new and an emerging field. The overarching aim of this review is to provide an update on recent literature focused on the use of saliva as a biological sample matrix in microfluidic devices. We will first cover the characteristics of saliva as a sample medium and then review microfluidic devices that are developed for the analysis of salivary biomarkers.
Collapse
Affiliation(s)
- Trey W Pittman
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Daniel Balazs Decsi
- Centre for Biomedical Technologies, School of Biomedical Sciences, Faculty of Health, QUT.,Griffith Institute for Drug Discover, Griffith University, Nathan, Australia
| | - Chamindie Punyadeera
- Griffith Institute for Drug Discover, Griffith University, Nathan, Australia.,Menzies Health Institute, Griffith University, Gold Coast, Australia.,Translational Research Institute, Woolloongabba, Australia
| | - Charles S Henry
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523, USA.,Metallurgy and Materials Science Research Institute, Chulalongkorn University, Soi Chula 12, Phayathai Rd., Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
48
|
Fu H, Sun L, Zhu J. Detection of Antibody versus Antigen, Optimal Option of Different Serological Assays Based Tests for COVID-19 Diagnosis: A Meta-Analysis. IRANIAN JOURNAL OF PUBLIC HEALTH 2023; 52:23-36. [PMID: 36824236 PMCID: PMC9941426 DOI: 10.18502/ijph.v52i1.11662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/11/2022] [Indexed: 01/19/2023]
Abstract
Background In this study, the diagnostic efficacy of antigen test and antibody test were assessed. Additionally, the difference of sensitivity, specificity, and diagnostic odds ratio were compared concerning efficacy of antibody test versus antigen test for Corona Virus Disease 2019 (COVID-19) diagnosis. Methods Online databases were searched for full-text publications and STATA software was used for data pooling and analysis before Sep 1st, 2022. Forrest plot was used to show the pooled sensitivity, specificity and diagnostic odds ratio. Combined receiver operating characteristic (ROC) curve was used to show the area of under curve of complex data. Results Overall, 25 studies were included. The sensitivity (0.68, 95% CI: 0.53-0.80) and specificity (0.99, 95% CI: 0.98-0.99) in antibody or antigen was calculated. The time point of test lead to heterogeneity. The area under curve (AUC) was 0.98 (95% CI: 0.96-0.99), and the diagnostic odds ratio (DOR) was 299.54 (95% CI: 135.61-661.64). Subgroup analysis indicated antibody test with sensitivity (0.59, 95% CI: 0.44-0.73) and specificity (0.98, 95% CI: 0.95-0.99) and antigen test with sensitivity of 0.77 (95% CI: 0.53-0.91) and specificity of 0.99 (95% CI: 0.98-1.00). Higher AUC and DOR were proved in antigen test. Conclusion The present study compared the efficacy of antibody test versus antigen test for COVID-19 diagnosis. Better diagnostic efficacy, lower heterogeneity, and less publication bias of rapid antigen testing was suggested in this study. This study would help us to make better strategy about choosing rapid and reliable testing method in diagnosis of the COVID-19 disease.
Collapse
Affiliation(s)
- Haiyan Fu
- Department of Clinical Laboratory, Yantaishan Hospital, Yantai 264001, Shandong Province, PR China
| | - Lin Sun
- Department of Clinical Laboratory, Yantaishan Hospital, Yantai 264001, Shandong Province, PR China
| | - Jingwei Zhu
- Department of Clinical Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong Province, PR China,Corresponding Author:
| |
Collapse
|
49
|
Schwob JM, Miauton A, Petrovic D, Perdrix J, Senn N, Gouveia A, Jaton K, Opota O, Maillard A, Minghelli G, Cornuz J, Greub G, Genton B, D’Acremont V. Antigen rapid tests, nasopharyngeal PCR and saliva PCR to detect SARS-CoV-2: A prospective comparative clinical trial. PLoS One 2023; 18:e0282150. [PMID: 36827328 PMCID: PMC9955963 DOI: 10.1371/journal.pone.0282150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 01/07/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Nasopharyngeal antigen Rapid Diagnostic Tests (RDTs), saliva RT-PCR and nasopharyngeal (NP) RT-PCR have shown different performance characteristics to detect patients infected by SARS-CoV-2, according to the viral load (VL)-and thus transmissibility. METHODS In October 2020, we conducted a prospective trial involving patients presenting at testing centres with symptoms of COVID-19. We compared detection rates and performance of RDT, saliva PCR and nasopharyngeal (NP) PCR, according to VL and symptoms duration. RESULTS Out of 949 patients enrolled, 928 patients had all three tests performed. Detection rates were 35.2% (95%CI 32.2-38.4%) by RDT, 39.8% (36.6-43.0%) by saliva PCR, 40.1% (36.9-43.3%) by NP PCR, and 41.5% (38.3-44.7%) by any test. For those with viral loads (VL) ≥106 copies/ml, detection rates were 30.3% (27.3-33.3), 31.4% (28.4-34.5), 31.5% (28.5-34.6), and 31.6% (28.6-34.7%) respectively. Sensitivity of RDT compared to NP PCR was 87.4% (83.6-90.6%) for all positive patients, 94.5% (91.5-96.7%) for those with VL≥105 and 96.5% (93.6-98.3%) for those with VL≥106. Sensitivity of STANDARD-Q®, Panbio™ and COVID-VIRO® Ag tests were 92.9% (86.4-96.9%), 86.1% (78.6-91.7%) and 84.1% (76.9-89.7%), respectively. For those with VL≥106, sensitivity was 96.6% (90.5-99.3%), 97.8% (92.1-99.7%) and 95.3% (89.4-98.5%) respectively. No patient with VL<104 was detected by RDT. Specificity of RDT was 100% (99.3-100%) compared to any PCR. RDT sensitivity was similar <4 days (87.8%, 83.5-91.3%) and ≥4 days (85.7%, 75.9-92.6%) after symptoms onset (p = 0.6). Sensitivity of saliva and NP PCR were 95.7% (93.1-97.5%) and 96.5% (94.1-98.1%), respectively, compared to the other PCR. CONCLUSIONS RDT results allow rapid identification of COVID cases with immediate isolation of most contagious individuals. RDT can thus be a game changer both in ambulatory care and community testing aimed at stopping transmission chains, and even more so in resource-constrained settings thanks to its very low price. When PCR is performed, saliva could replace NP swabbing. TRIAL REGISTRATION ClinicalTrial.gov Identifier: NCT04613310 (03/11/2020).
Collapse
Affiliation(s)
- Jean-Marc Schwob
- Department of Policlinics, Centre for Primary Care and Public Health (Unisanté), Lausanne, Switzerland
- * E-mail:
| | - Alix Miauton
- Department of Policlinics, Centre for Primary Care and Public Health (Unisanté), Lausanne, Switzerland
| | - Dusan Petrovic
- Department of Epidemiology and Health Systems, Centre for Primary Care and Public Health (Unisanté), Lausanne, Switzerland
| | - Jean Perdrix
- Department of Policlinics, Centre for Primary Care and Public Health (Unisanté), Lausanne, Switzerland
| | - Nicolas Senn
- Department of Policlinics, Centre for Primary Care and Public Health (Unisanté), Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
| | - Alexandre Gouveia
- Department of Policlinics, Centre for Primary Care and Public Health (Unisanté), Lausanne, Switzerland
| | - Katia Jaton
- University of Lausanne, Lausanne, Switzerland
- Institute of Microbiology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Onya Opota
- University of Lausanne, Lausanne, Switzerland
- Institute of Microbiology, University Hospital of Lausanne, Lausanne, Switzerland
| | | | | | - Jacques Cornuz
- Department of Policlinics, Centre for Primary Care and Public Health (Unisanté), Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
| | - Gilbert Greub
- University of Lausanne, Lausanne, Switzerland
- Institute of Microbiology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Blaise Genton
- Department of Policlinics, Centre for Primary Care and Public Health (Unisanté), Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
- Department of Training, Research and Innovation, Centre for Primary Care and Public Health (Unisanté), Lausanne, Switzerland
| | - Valérie D’Acremont
- Department of Policlinics, Centre for Primary Care and Public Health (Unisanté), Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
- Department of Training, Research and Innovation, Centre for Primary Care and Public Health (Unisanté), Lausanne, Switzerland
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| |
Collapse
|
50
|
Farsaeivahid N, Grenier C, Nazarian S, Wang ML. A Rapid Label-Free Disposable Electrochemical Salivary Point-of-Care Sensor for SARS-CoV-2 Detection and Quantification. SENSORS (BASEL, SWITZERLAND) 2022; 23:433. [PMID: 36617031 PMCID: PMC9823438 DOI: 10.3390/s23010433] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 05/24/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has created an urgent need for accurate early diagnosis and monitoring. A label-free rapid electrochemical point-of-care (POC) biosensor for SARS-CoV-2 detection in human saliva is reported here to help address the shortcomings of traditional nucleic acid amplification methods and give a quantitative assessment of the viral load to track infection status anywhere, using disposable electrochemical sensor chips. A new chemical construct of gold nanoparticles (GNp) and thionine (Th) are immobilized on carboxylic acid functionalized carbon nanotubes (SWCNT-COOH) for high-performance biosensing. The sensor uses saliva with a one-step pretreatment and simple testing procedure as an analytical medium due to the user-friendly and non-invasive nature of its procurement from patients. The sensor has a response time of 5 min with a limit of detection (LOD) reaching 200 and 500 pM for the freely suspended spike (S) protein in phosphate buffer saline (PBS) and human saliva, respectively. The sensor's performance was also proven for detecting a COVID-19 pseudovirus in an electrolyte solution with a LOD of 106 copies/mL. The results demonstrate that the optimized POC sensor developed in this work is a promising device for the label-free electrochemical biosensing detection of SARS-CoV-2 and different species of viruses.
Collapse
Affiliation(s)
- Nadia Farsaeivahid
- Interdisciplinary Engineering Program, Northeastern University, Boston, MA 02115, USA
| | - Christian Grenier
- Interdisciplinary Engineering Program, Northeastern University, Boston, MA 02115, USA
| | - Sheyda Nazarian
- Interdisciplinary Engineering Program, Northeastern University, Boston, MA 02115, USA
| | - Ming L. Wang
- Civil and Environmental Engineering Department, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|