Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Bio Protoc ; 11(9): e4005, 2021 May 05.
Article in English | MEDLINE | ID: covidwho-2326923

ABSTRACT

The COVID-19 pandemic requires mass screening to identify those infected for isolation and quarantine. Individually screening large populations for the novel pathogen, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), is costly and requires a lot of resources. Sample pooling methods improve the efficiency of mass screening and consume less reagents by increasing the capacity of testing and reducing the number of experiments performed, and are therefore especially suitable for under-developed countries with limited resources. Here, we propose a simple, reliable pooling strategy for COVID-19 testing using clinical nasopharyngeal (NP) and/or oropharyngeal (OP) swabs. The strategy includes the pooling of 10 NP/OP swabs for extraction and subsequent testing via quantitative real-time reverse transcription polymerase chain reaction (RT-qPCR), and may also be applied to the screening of other pathogens.

2.
International Journal of Parallel, Emergent and Distributed Systems ; 2023.
Article in English | Scopus | ID: covidwho-2268733

ABSTRACT

It is well and widely known that sample pooling could provide an effective and efficient way for fast coronavirus testing among massive asymptomatic individuals. The method of multi-level acceleration for asymptomatic COVID-19 screening has been introduced, and for one and two levels, the optimal group sizes have been obtained. However, there are still multiple challenges. First, it is not clear how to find the optimal group sizes for three or more levels. Second, there is lack of closed-form expressions for the optimal group sizes for two or more levels. Third, it is not clear how to determine the optimal number of levels. And last, it is not known what the maximum achievable speedup is. The motivation of this paper is to address all the above challenges. The optimization of a hierarchical pooling strategy includes its number of levels and the group size of each level. In this paper, based on multi-variable optimization and Taylor approximation, we are able to derive closed-form expressions for the optimal number of levels (Formula presented.), the optimal group sizes (Formula presented.), (Formula presented.),…, (Formula presented.), and the maximum possible speedup of a hierarchical pooling strategy of (Formula presented.), where (Formula presented.) is the fraction of infected people. The above speedup is nearly a linear function of the reciprocal of (Formula presented.), in the sense that it is asymptotically greater than any sub-linear function (Formula presented.) of the reciprocal of (Formula presented.) for any small (Formula presented.). Using the results in this paper, we can quickly and easily predict the performance of an optimal hierarchical pooling strategy. For instance, if the fraction of infected people is 0.0001, an 8-level hierarchical pooling strategy can achieve speedup of nearly 400. © 2023 Informa UK Limited, trading as Taylor & Francis Group.

3.
VIEW ; 3(4), 2022.
Article in English | Scopus | ID: covidwho-2282135

ABSTRACT

The outbreak of coronavirus disease 2019 (COVID-19, caused by SARS-Cov-2) is a big challenge for global health systems and the economy. Rapid and accurate tests are crucial at early stages of this pandemic. Reverse transcription-quantitative real-time polymerase chain reaction is the current gold standard method for detection of SARS-Cov-2. It is impractical and costly to test individuals in large-scale population screens, especially in low- and middle-income countries due to their shortage of nucleic acid testing reagents and skilled staff. Accordingly, sample pooling, such as for blood screening for syphilis, is now widely applied to COVID-19. In this paper, we survey and review several different pooled-sample testing strategies, based on their group size, prevalence, testing number, and sensitivity, and we discuss their efficiency in terms of reducing cost and saving time while ensuring sensitivity. © 2022 The Authors. VIEW published by Shanghai Fuji Technology Consulting Co., Ltd, authorized by Professional Community of Experimental Medicine, National Association of Health Industry and Enterprise Management (PCEM) and John Wiley & Sons Australia, Ltd.

4.
Front Microbiol ; 13: 957957, 2022.
Article in English | MEDLINE | ID: covidwho-1987529

ABSTRACT

COVID-19 is a life-threatening multisistemic infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Infection control relies on timely identification and isolation of infected people who can alberg the virus for up to 14 days, providing important opportunities for undetected transmission. This note describes the application of rRT-PCR test for simpler, faster and less invasive monitoring of SARS-CoV-2 infection using pooling strategy of samples. Seventeen positive patients were provided with sterile dry swabs and asked to self-collected 2 nasal specimens (#NS1 and #NS2). The #NS1 was individually placed in a single tube and the #NS2 was placed in another tube together with 19 NSs collected from 19 negative patients. Both tubes were then tested with conventional molecular rRT-PCR and the strength of pooling nasal testing was compared with the molecular test performed on the single NS of each positive patient. The pooling strategy detected SARS-CoV-2 RNA to a similar extent to the single test, even when Ct value is on average high (Ct 37-38), confirming that test sensibility is not substantially affected even if the pool contains only one low viral load positive sample. Furthermore, the pooling strategy have benefits for SARS-CoV-2 routinary monitoring of groups in regions with a low SARS-CoV-2 prevalence.

5.
Acad Pathol ; 8: 23742895211023948, 2021.
Article in English | MEDLINE | ID: covidwho-1304387

ABSTRACT

The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2, led to unprecedented demands assigned to clinical diagnostic laboratories worldwide, forcing them to make significant changes to their regular workflow as they adapted to new diagnostic tests and sample volumes. Herein, we summarize the modifications/adaptation the laboratory had to exercise to cope with rapidly evolving situations in the current pandemic. In the first phase of the pandemic, the laboratory validated 2 reverse transcription polymerase chain reaction-based assays to test ∼1000 samples/day and rapidly modified procedures and validated various preanalytical and analytical steps to overcome the supply chain constraints that would have otherwise derailed testing efforts. Further, the pooling strategy was validated for wide-scale population screening using nasopharyngeal swab samples and saliva samples. The translational research arm of the laboratory pursued several initiatives to understand the variable clinical manifestations that this virus presented in the population. The phylogenetic evolution of the virus was investigated using next-generation sequencing technology. The laboratory has initiated the formation of a consortium that includes groups investigating genomes at the level of large structural variants, using genome optical mapping via this collaborative global effort. This article summarizes our journey as the laboratory has sought to adapt and continue to positively contribute to the unprecedented demands and challenges of this rapidly evolving pandemic.

6.
Int J Infect Dis ; 105: 83-90, 2021 Apr.
Article in English | MEDLINE | ID: covidwho-1077929

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the feasibility of saliva sampling as a non-invasive and safer tool to detect severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and to compare its reproducibility and sensitivity with nasopharyngeal swab samples (NPS). The use of sample pools was also investigated. METHODS: A total of 2107 paired samples were collected from asymptomatic healthcare and office workers in Mexico City. Sixty of these samples were also analyzed in two other independent laboratories for concordance analysis. Sample processing and analysis of virus genetic material were performed according to standard protocols described elsewhere. A pooling analysis was performed by analyzing the saliva pool and the individual pool components. RESULTS: The concordance between NPS and saliva results was 95.2% (kappa 0.727, p = 0.0001) and 97.9% without considering inconclusive results (kappa 0.852, p = 0.0001). Saliva had a lower number of inconclusive results than NPS (0.9% vs 1.9%). Furthermore, saliva showed a significantly higher concentration of both total RNA and viral copies than NPS. Comparison of our results with those of the other two laboratories showed 100% and 97% concordance. Saliva samples are stable without the use of any preservative, and a positive SARS-CoV-2 sample can be detected 5, 10, and 15 days after collection when the sample is stored at 4 °C. CONCLUSIONS: The study results indicate that saliva is as effective as NPS for the identification of SARS-CoV-2-infected asymptomatic patients. Sample pooling facilitates the analysis of a larger number of samples, with the benefit of cost reduction.


Subject(s)
COVID-19/diagnosis , SARS-CoV-2/isolation & purification , Saliva/virology , Cross-Sectional Studies , Humans , Nasopharynx/virology , Reproducibility of Results , Specimen Handling
7.
J Med Virol ; 93(5): 2805-2809, 2021 05.
Article in English | MEDLINE | ID: covidwho-891891

ABSTRACT

The emerging pandemic of coronavirus disease 2019 (COVID-19) has affected over 200 countries and resulted in a shortage of diagnostic resources globally. Rapid diagnosis of COVID-19 is vital to control the spreading of the disease, which, however, is challenged by limited detection capacity and low detection efficiency in many parts of the world. The pooling test may offer an economical and effective approach to increase the virus testing capacity of medical laboratories without requiring more laboratory resources such as laboratory workers, testing reagents, and equipment. In this study, the sample pools of 6 and 10 were detected by a real-time reverse transcription-polymerase chain reaction assay targeting ORF1ab and N genes of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Each pool consisted of five or nine negative SARS-CoV-2 samples and one positive counterpart with varying viral loads. Two different strategies of sample pooling were investigated and the results were compared comprehensively. One approach was to pool the viral transport medium of the samples in the laboratory, and the other was to pool swab samples during the collection process. For swab pooling strategy, qualitative results of SARS-CoV-2 RNA, specific tests of ORF1ab and N genes, remained stable over the different pool sizes. Together, this study demonstrates that the swab pooling strategy may serve as an effective and economical approach for screening SARS-CoV-2 infections in large populations, especially in countries and regions where medical resources are limited during the pandemic and may thus be potential for clinical laboratory applications.


Subject(s)
COVID-19 Testing/methods , COVID-19/diagnosis , RNA, Viral/isolation & purification , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , COVID-19 Nucleic Acid Testing/methods , Coronavirus Nucleocapsid Proteins/genetics , Diagnostic Tests, Routine/methods , Humans , Mass Screening/methods , Phosphoproteins/genetics , Polyproteins/genetics , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction/methods , Specimen Handling/methods , Viral Load , Viral Proteins/genetics
8.
Diagnostics (Basel) ; 10(7)2020 Jul 11.
Article in English | MEDLINE | ID: covidwho-646822

ABSTRACT

Due to the current pandemic, a global shortage of reagents has drawn interest in developing alternatives to increase the number of coronavirus tests. One such alternative is sample pooling. We compared commercial kits that are used in COVID-19 diagnostics in terms of their sensitivity and feasibility for use in pooling. In this preliminary study, we showed that pooling of up to 80 samples did not affect the efficacy of the kits. Additionally, the RNA-dependent RNA polymerase (RdRp) gene is a more suitable target in pooled samples than the envelope (E) gene. This approach could provide an easy method of screening a large number of samples and help adjust different governmental regulations.

SELECTION OF CITATIONS
SEARCH DETAIL