Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 29
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.
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.

3.
Comp Clin Path ; 32(3): 375-381, 2023.
Article in English | MEDLINE | ID: covidwho-2238273

ABSTRACT

Sample pooling testing for SARS-COV-2 can be an effective tool in COVID-19 screening when resources are limited, yet it is important to assess the performance before implementation as pooling has its limitations. Our objective was to assess the efficacy of pooling samples for coronavirus 2019 (COVID-19) compared to an individual analysis by using commercial platforms for nucleic acid testing. A total of 2200 nasopharyngeal swabs for SARS-COV-2 were tested individually and in pools of 4, 8, and 10. The cycle threshold (Ct) values of the positive pooled samples were compared to their corresponding individual positive samples. In pool size 10 samples, an estimated increase of 3-Ct was obtained, which led to false negative results in low viral load positive samples. Pooling SARS COV-2 samples is an effective strategy of screening to increase laboratories' capacity and reduce costs without affecting diagnostic performance. A pool size of 8 is recommended.

4.
J Nepal Health Res Counc ; 20(2): 372-376, 2022 Nov 02.
Article in English | MEDLINE | ID: covidwho-2206052

ABSTRACT

BACKGROUND: Rapid detection of Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) by real-time polymerase chain reaction (RT-PCR) is the most reliable method used worldwide. Although the incidence of the disease has increased globally, the limited availability of PCR kits has become the major bottleneck for the diagnosis of COVID positive patients. METHODS: Random samples were pooled for two months in group of two-five and tested for SARS-CoV-2. If the pool was negative, all individuals in the pool were reported negative. If the pool was positive, then the individual samples were retested to identify the positive individual. RESULTS: The mean cycle threshold (Ct) value of pooled samples was not significantly different with that of individual samples for N, ORF-1ab and E genes. Also, pooling saved more than 60% of reagents, time and effort, workforce and cost. CONCLUSIONS: In this study, the positivity rate was around 5% and saving of reagent, cost, time and manpower was more than 60%.


Subject(s)
COVID-19 , Humans , COVID-19/diagnosis , COVID-19/epidemiology , SARS-CoV-2/genetics , Real-Time Polymerase Chain Reaction/methods , Cost-Benefit Analysis , Nepal , Sensitivity and Specificity , Reverse Transcriptase Polymerase Chain Reaction
5.
Front Public Health ; 10: 1053873, 2022.
Article in English | MEDLINE | ID: covidwho-2199533

ABSTRACT

This study aims to propose a pooling approach to simulate the compulsory universal RT-PCR test in Hong Kong and explore the feasibility of implementing the pooling method on a household basis. The mathematical model is initially verified, and then the simulation is performed under different prevalence rates and pooled sizes. The simulated population is based in Hong Kong. The simulation included 10,000,000 swab samples, with a representative distribution of populations in Hong Kong. The samples were grouped into a batch size of 20. If the entire batch is positive, then the group is further divided into an identical group size of 10 for re-testing. Different combinations of mini-group sizes were also investigated. The proposed pooling method was extended to a household basis. A representative from each household is required to perform the RT-PCR test. Results of the simulation replications, indicate a significant reduction (p < 0.001) of 83.62, 64.18, and 48.46% in the testing volume for prevalence rate 1, 3, and 5%, respectively. Combined with the household-based pooling approach, the total number of RT-PCR is 437,304, 956,133, and 1,375,795 for prevalence rates 1, 3, and 5%, respectively. The household-based pooling strategy showed efficiency when the prevalence rates in the population were low. This pooling strategy can rapidly screen people in high-risk groups for COVID-19 infections and quarantine those who test positive, even when time and resources for testing are limited.


Subject(s)
COVID-19 , Humans , COVID-19/diagnosis , COVID-19/epidemiology , Hong Kong/epidemiology , COVID-19 Testing , Specimen Handling , Prevalence
6.
View ; 3(4), 2022.
Article in English | ProQuest Central | ID: covidwho-1958862

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.

7.
Diagnostics (Basel) ; 12(6)2022 Jun 06.
Article in English | MEDLINE | ID: covidwho-1884054

ABSTRACT

As COVID-19 transmission control measures are gradually being lifted, a sensitive and rapid diagnostic method for large-scale screening could prove essential for monitoring population infection rates. However, many rapid workflows for SARS-CoV-2 detection and diagnosis are not amenable to the analysis of large-volume samples. Previously, our group demonstrated a technique for SARS-CoV-2 nanoparticle-facilitated enrichment and enzymatic lysis from clinical samples in under 10 min. Here, this sample preparation strategy was applied to pooled samples originating from nasopharyngeal (NP) swabs eluted in viral transport medium (VTM) and saliva samples diluted up to 1:100. This preparation method was coupled with conventional RT-PCR on gold-standard instrumentation for proof-of-concept. Additionally, real-time PCR analysis was conducted using an in-house, ultra-rapid real-time microfluidic instrument paired with an experimentally optimized rapid protocol. Following pooling and extraction from clinical samples, average cycle threshold (CT) values from resultant eluates generally increased as the pooling dilution factor increased; further, results from a double-blind study demonstrated 100% concordance with clinical values. In addition, preliminary data obtained from amplification of eluates prepared by this technique and analyzed using our portable, ultra-rapid real-time microfluidic PCR amplification instrument showed progress toward a streamlined method for rapid SARS-CoV-2 analysis from pooled samples.

8.
Pathology ; 54(4): 466-471, 2022 Jun.
Article in English | MEDLINE | ID: covidwho-1799763

ABSTRACT

During the COVID-19 pandemic, sample pooling has proven an effective strategy to overcome the limitations of reagent shortages and expand laboratory testing capacity. The inclusion of influenza and respiratory syncytial virus (RSV) in a multiplex tandem PCR platform with SARS-CoV-2 provides useful diagnostic and infection control information. This study aimed to evaluate the performance of the influenza and RSV targets in the AusDiagnostics SARS-CoV-2, Influenza and RSV 8-well assay, including the effect of pooling samples on target detection. RSV target detection in clinical samples was compared to the Cepheid Xpert Xpress Flu/RSV assay as a reference standard. Samples were then tested in pools of four and detection rates were compared. Owing to the unavailability of clinical samples for influenza, only the effect of sample pooling on simulated samples was evaluated for these targets. RSV was detected in neat clinical samples with a positive percent agreement (PPA) of 100% and negative percent agreement (NPA) of 99.5% compared to the reference standard, demonstrating 99.7% agreement. This study demonstrates that sample pooling by four increases the average Ct value by 2.24, 2.29, 2.20 and 1.91 cycles for the target's influenza A, influenza A typing, influenza B and RSV, respectively. The commercial AusDiagnostics SARS-CoV-2, Influenza and RSV 8-well assay was able to detect influenza and RSV at an intermediate concentration within the limit of detection of the assay. Further studies to explore the applicability of sample pooling at the lower limit of detection of the assay is needed. Nevertheless, sample pooling has shown to be a viable strategy to increase testing throughput and reduce reagent usage. In addition, the multiplexed platform targeting various respiratory viruses assists with public health and infection control responses, clinical care, and patient management.


Subject(s)
COVID-19 , Influenza A virus , Influenza, Human , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , COVID-19/diagnosis , Humans , Influenza A virus/genetics , Influenza B virus/genetics , Influenza, Human/diagnosis , Molecular Diagnostic Techniques , Nasopharynx , Pandemics , Respiratory Syncytial Virus Infections/diagnosis , Respiratory Syncytial Virus, Human/genetics , SARS-CoV-2 , Sensitivity and Specificity
9.
Ieee Open Journal of the Computer Society ; 1:276-284, 2020.
Article in English | Web of Science | ID: covidwho-1583787

ABSTRACT

Testing has been a major factor that limits our response to the COVID-19 pandemic. The method of sample pooling and group test has recently been introduced and adopted. However, it is still not clearly known how to determine the appropriate group size. In this paper, we treat asymptomatic COVID-19 screening acceleration as an optimization problem, and solve the problem using an analytical approach and an algorithmic procedure. We develop a two-level hierarchical pooling strategy for accelerating asymptomatic COVID-19 screening. In the first level, a population is divided into groups, which results in inter-group acceleration. In the second level, a group is divided into subgroups, which results in intra-group and intersubgroup acceleration. By using our analytical methods and numerical algorithms, we determine the optimal group size and the optimal subgroup size, which minimize the total number of tests, maximize the speedup of the hierarchical pooling strategy, and minimize both time and cost of testing. It is discovered that the optimal group size and the optimal subgroup size are determined by the fraction of infected people. Furthermore, the optimal group size, the optimal subgroup size, and the achieved speedup grow sublinearly with the reciprocal of the fraction of infected people. Our research has important social implications and financial impacts. For example, if the fraction of infected people is 0.01, by using group size of 25 and subgroup size of 5, we can achieve speedup of at least 11, which means that months of testing time can be reduced to days, and over 91% of the testing cost can be saved. Such results have not been available in the known literature. The paper makes significant progress and great advance in pooling strategy optimization for accelerating asymptomatic COVID-19 screening, and represents the contribution of computer science to the global pandemic.

10.
Int J Environ Res Public Health ; 18(17)2021 09 03.
Article in English | MEDLINE | ID: covidwho-1390638

ABSTRACT

Reliability, accuracy, and timeliness of diagnostic testing for SARS-CoV-2 infection have allowed adequate public health management of the disease, thus notably helping the timely mapping of viral spread within the community. Furthermore, the most vulnerable populations, such as people with intellectual disability and dementia, represent a high-risk group across multiple dimensions, including a higher prevalence of pre-existing conditions, lower health maintenance, and a propensity for rapid community spread. This led to an urgent need for reliable in-house rapid testing to be performed prior to hospital admission. In the present study, we describe a pooling procedure in which oropharyngeal and nasopharyngeal swabs for SARS-CoV-2 detection (performed prior to hospital admission using rapid RT-PCR assay) are pooled together at the time of sample collection. Sample pooling (groups of 2-4 samples per tube) allowed us to significantly reduce response times, consumables, and personnel costs while maintaining the same test sensitivity.


Subject(s)
COVID-19 , Intellectual Disability , Hospitals , Humans , Intellectual Disability/diagnosis , Intellectual Disability/epidemiology , Real-Time Polymerase Chain Reaction , Reproducibility of Results , SARS-CoV-2 , Sensitivity and Specificity
11.
J Clin Virol ; 141: 104895, 2021 08.
Article in English | MEDLINE | ID: covidwho-1293947

ABSTRACT

BACKGROUND: Worldwide demand for SARS-CoV-2 RT-PCR testing is still high as testing remains central to follow the disease spread and vaccine efficacy. Group testing has been proposed as a solution to expand testing capabilities but sensitivity concerns may limit its impact on the management of the pandemic. Digital PCR (RT-dPCR) has been shown to be highly sensitive and could help by providing larger testing capabilities without compromising sensitivity. METHODS: We implemented RT-dPCR based COVID-19 group testing on a commercially available system and assay (naica® system from Stilla Technologies) and investigated the sensitivity of the method in real life conditions of a university hospital in Paris, France, in May 2020. We tested the protocol in a direct comparison with reference RT-PCR testing on 448 samples split into groups of 8, 16 and 32 samples for RT-dPCR analysis. RESULTS: Individual RT-PCR testing identified 25/448 positive samples. Using 56 groups of 8, RT-dPCR identified 23 groups as positive, corresponding to 26 positive samples by individual PCR (positive percentage agreement 95.2% [95% confidence interval: 76.2-99.9%]) and including 2 samples not detected by individual RT-PCR but confirmed positive by further investigation. 15 of 28 groups of 16 tested positive, corresponding to 25 positive samples by individual PCR (positive percentage agreement 87.5% [95% confidence interval: 61.7-98.4%]). 14 groups of 32 were fully concordant with individual PCR testing but will need to be confirmed on larger datasets. CONCLUSIONS: Our proposed approach of group testing by digital PCR has similar diagnostic sensitivity compared to individual RT-PCR testing for group up to 16 samples. This approach reduces the quantity of reagent needed by up to 80% while reducing costs and increasing capabilities of testing up to 10-fold.


Subject(s)
COVID-19 , SARS-CoV-2 , Hospitals , Humans , Pandemics , Polymerase Chain Reaction , Sensitivity and Specificity
12.
Diagnostics (Basel) ; 11(7)2021 Jun 26.
Article in English | MEDLINE | ID: covidwho-1288830

ABSTRACT

Since the beginning of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic, it has been clear that testing large groups of the population was the key to stem infection and prevent the effects of the coronavirus disease of 2019, mostly among sensitive patients. On the other hand, time and cost-sustainability of virus detection by molecular analysis such as reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) may be a major issue if testing is extended to large communities, mainly asymptomatic large communities. In this context, sample-pooling and test grouping could offer an effective solution. Here we report the screening on 1195 oral-nasopharyngeal swabs collected from students and staff of the Università degli Studi del Sannio (University of Sannio, Benevento, Campania, Italy) and analyzed by an in-house developed multiplex RT-qPCR for SARS-CoV-2 detection through a simple monodimensional sample pooling strategy. Overall, 400 distinct pools were generated and, within 24 h after swab collection, five positive samples were identified. Out of them, four were confirmed by using a commercially available kit suitable for in vitro diagnostic use (IVD). High accuracy, sensitivity and specificity were also determined by comparing our results with a reference IVD assay for all deconvoluted samples. Overall, we conducted 463 analyses instead of 1195, reducing testing resources by more than 60% without lengthening diagnosis time and without significant losses in sensitivity, suggesting that our strategy was successful in recognizing positive cases in a community of asymptomatic individuals with minor requirements of reagents and time when compared to normal testing procedures.

13.
J Clin Lab Anal ; 35(8): e23876, 2021 Aug.
Article in English | MEDLINE | ID: covidwho-1270480

ABSTRACT

BACKGROUND: Pooling of samples for SARS-CoV-2 testing in low-prevalence settings has been used as an effective strategy to expand testing capacity and mitigate challenges with the shortage of supplies. We evaluated two automated molecular test systems for the detection of SARS-CoV-2 RNA in pooled specimens. METHODS: Pooled nasopharyngeal and saliva specimens were tested by Qiagen QIAstat-Dx Respiratory SARS-CoV-2 Panel (QIAstat) or Cepheid Xpert Xpress SARS-CoV-2 (Xpert), and the results were compared to that of standard RT-qPCR tests without pooling. RESULTS: In nasopharyngeal specimens, the sensitivity/specificity of the pool testing approach, with 5 and 10 specimens per pool, were 77%/100% (n = 105) and 74.1%/100% (n = 260) by QIAstat, and 97.1%/100% (n = 250) and 100%/99.5% (n = 200) by Xpert, respectively. Pool testing of saliva (10 specimens per pool; n = 150) by Xpert resulted in 87.5% sensitivity and 99.3% specificity compared to individual tests. Pool size of 5 or 10 specimens did not significantly affect the difference of RT-qPCR cycle threshold (CT ) from standard testing. RT-qPCR CT values obtained with pool testing by both QIAstat and Xpert were positively correlated with that of individual testing (Pearson's correlation coefficient r = 0.85 to 0.99, p < 0.05). However, the CT values from Xpert were significantly stronger (p < 0.01, paired t test) than that of QIAstat in a subset of SARS-CoV-2 positive specimens, with mean differences of -4.3 ± 2.43 and -4.6 ± 2 for individual and pooled tests, respectively. CONCLUSION: Our results suggest that Xpert SARS-CoV-2 can be utilized for pooled sample testing for COVID-19 screening in low-prevalence settings providing significant cost savings and improving throughput without affecting test quality.


Subject(s)
COVID-19 Testing/methods , Nasopharynx/virology , Saliva/virology , Automation, Laboratory , COVID-19 Nucleic Acid Testing/methods , Humans , Molecular Diagnostic Techniques/methods , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity
14.
Viruses ; 13(5)2021 05 13.
Article in English | MEDLINE | ID: covidwho-1227073

ABSTRACT

The primary approach to controlling the spread of the pandemic SARS-CoV-2 is to diagnose and isolate the infected people quickly. Our paper aimed to investigate the efficiency and the reliability of a hierarchical pooling approach for large-scale PCR testing for SARS-CoV-2 diagnosis. To identify the best conditions for the pooling approach for SARS-CoV-2 diagnosis by RT-qPCR, we investigated four manual methods for both RNA extraction and PCR assessment targeting one or more of the RdRp, N, S, and ORF1a genes, by using two PCR devices and an automated flux for SARS-CoV-2 detection. We determined the most efficient and accurate diagnostic assay, taking into account multiple parameters. The optimal pool size calculation included the prevalence of SARS-CoV-2, the assay sensitivity of 95%, an assay specificity of 100%, and a range of pool sizes of 5 to 15 samples. Our investigation revealed that the most efficient and accurate procedure for detecting the SARS-CoV-2 has a detection limit of 2.5 copies/PCR reaction. This pooling approach proved to be efficient and accurate in detecting SARS-CoV-2 for all samples with individual quantification cycle (Cq) values lower than 35, accounting for more than 94% of all positive specimens. Our data could serve as a comprehensive practical guide for SARS-CoV-2 diagnostic centers planning to address such a pooling strategy.


Subject(s)
COVID-19 Testing/methods , COVID-19/diagnosis , SARS-CoV-2/genetics , COVID-19/blood , COVID-19/genetics , High-Throughput Screening Assays/methods , Humans , Pandemics/prevention & control , RNA, Viral/blood , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction/methods , Reproducibility of Results , SARS-CoV-2/pathogenicity , Sensitivity and Specificity , Specimen Handling/methods
15.
J Med Virol ; 93(4): 2281-2286, 2021 04.
Article in English | MEDLINE | ID: covidwho-1217380

ABSTRACT

There are more than 350 real-time polymerase chain reaction (RT-PCR) coronavirus disease-2019 (COVID-19) testing kits commercially available but these kits have not been evaluated for pooled sample testing. Thus, this study was planned to compare and evaluate seven commercially available kits for pooled samples testing. Diagnostic accuracy of (1) TRUPCR SARS-CoV-2 Kit (Black Bio), (2) TaqPath RT-PCR COVID-19 Kit (Thermo Fisher Scientific), (3) Allplex 2019-nCOV Assay (Seegene), (4) Patho detect COVID-19 PCR kit (My Lab), (5) LabGun COVID-19 RT-PCR Kit (Lab Genomics, Korea), (6) Fosun COVID-19 RT-PCR detection kit (Fosun Ltd.), (7) Real-time Fluorescent RT-PCR kit for SARS CoV-2 (BGI) was evaluated on precharacterised 40 positive and 10 negative COVID-19 sample pools. All seven kits detected all sample pools with low Ct values (<30); while testing weak positive pooled samples with high Ct value (>30); the TRUPCR Kit, TaqPath Kit, Allplex Assay, and BGI RT-PCR kit showed 100% sensitivity, specificity, and accuracy. However, the Fosun kit, LabGun Kit, and Patho detect kit could detect only 90%, 85%, and 75% of weakly positive samples, respectively. We conclude that all seven commercially available RT-PCR kits included in this study can be used for routine molecular diagnosis of COVID-19. However, regarding performing pooled sample testing, it might be advisable to use those kits that performed best regarding positive identification in samples' pool, that is TRUPCR SARS-CoV-2 Kit, TaqPath RT-PCR COVID-19 Kit, Allplex 2019-nCOV Assay, and BGI Real-time RT-PCR kit for detecting SARS CoV-2.


Subject(s)
COVID-19 Testing/methods , COVID-19/diagnosis , Reagent Kits, Diagnostic , Real-Time Polymerase Chain Reaction/methods , SARS-CoV-2/isolation & purification , COVID-19/epidemiology , Clinical Laboratory Techniques , Humans , India/epidemiology , Prospective Studies , RNA, Viral/analysis , RNA, Viral/genetics , Republic of Korea , SARS-CoV-2/genetics , Sensitivity and Specificity , World Health Organization
16.
Biom J ; 63(6): 1272-1289, 2021 08.
Article in English | MEDLINE | ID: covidwho-1191268

ABSTRACT

We propose a mathematical model based on probability theory to optimize COVID-19 testing by a multistep batch testing approach with variable batch sizes. This model and simulation tool dramatically increase the efficiency and efficacy of the tests in a large population at a low cost, particularly when the infection rate is low. The proposed method combines statistical modeling with numerical methods to solve nonlinear equations and obtain optimal batch sizes at each step of tests, with the flexibility to incorporate geographic and demographic information. In theory, this method substantially improves the false positive rate and positive predictive value as well. We also conducted a Monte Carlo simulation to verify this theory. Our simulation results show that our method significantly reduces the false negative rate. More accurate assessment can be made if the dilution effect or other practical factors are taken into consideration. The proposed method will be particularly useful for the early detection of infectious diseases and prevention of future pandemics. The proposed work will have broader impacts on medical testing for contagious diseases in general.


Subject(s)
COVID-19 , Communicable Diseases , COVID-19/diagnosis , COVID-19 Testing , Communicable Diseases/diagnosis , Communicable Diseases/epidemiology , Computer Simulation , Humans , Pandemics , SARS-CoV-2
17.
BMC Infect Dis ; 21(1): 360, 2021 Apr 17.
Article in English | MEDLINE | ID: covidwho-1190060

ABSTRACT

BACKGROUND: The current pandemic of the SARS-CoV-2 virus, widely known as COVID-19, has affected millions of people around the world. The World Health Organization (WHO) has recommended vigorous testing to differentiate SARS-CoV-2 from other respiratory infections to aid in guiding appropriate care and management. Situations like this have demanded robust testing strategies and pooled testing of samples for SARS-CoV-2 virus has provided the solution to mass screening of people for COVID-19. A pooled testing strategy can be very effective in testing when resources are limited, yet it comes with its own limitations. These benefits and limitations need critical consideration when it comes to testing highly infectious diseases like COVID-19. METHODS: This study evaluated the pooled testing of nasopharyngeal swabs for SARS-COV-2 by comparing the sensitivity of individual sample testing with 4-and 8-pool sample testing. Median cycle threshold (Ct) values were compared, and the precision of pooled testing was assessed through an inter- and intra-assay of pooled samples. Coefficient of variance was calculated for inter- and intra-assay variability. RESULTS: The sensitivity becomes considerably lower when the samples are pooled. There is a high percentage of false negative reports with larger sample pool size and when the patient viral load is low or weak positive samples. High variability was seen in the intra- and inter-assay, especially among weak positive samples and when more number of samples are pooled together. CONCLUSION: As COVID - 19 infection numbers and need for testing remain high, we must meticulously evaluate the testing strategy for each country depending on its testing capacity, infrastructure, economic strength, and need to determine the optimal balance on the cost-effective strategy of resource saving and risk/ cost of missing positive patients.


Subject(s)
COVID-19 Testing/methods , COVID-19/diagnosis , Mass Screening/methods , Clinical Laboratory Techniques , Humans , Pandemics , SARS-CoV-2 , Sensitivity and Specificity , Viral Load
18.
J Med Virol ; 93(7): 4508-4515, 2021 Jul.
Article in English | MEDLINE | ID: covidwho-1156880

ABSTRACT

Early detection of the severe acute respiratory syndrome coronavirus 2 infection can decrease the spread of the disease and provide therapeutic options promptly in affected individuals. However, the diagnosis by reverse-transcription polymerase chain reaction is costly and time-consuming. Several methods of group testing have been developed to overcome this problem. The proposed strategy offers optimization of group testing according to the available resources by decreasing not only the number of the assays but also the turnaround time. The initial classification of the samples would be done according to the intention of testing defined as diagnostic or screening/surveillance, achieving the best possible homogeneity. The proposed stratification of pooling is based on branching (divisions) and depth (levels of re-pooling) of the original group in association with the estimated probability of a positive sample. The dilutional effect of the grouped samples has also been considered. The margins of minimum and maximum conservation of assays of pooled specimens are calculated and the optimum strategy can be selected in association with the probability of positive samples in the original group. This algorithm intends to be a useful tool for group testing offering a choice of strategies according to the requirements.


Subject(s)
COVID-19 Testing/methods , COVID-19/diagnosis , SARS-CoV-2/genetics , Specimen Handling/methods , Algorithms , Early Diagnosis , Humans , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity
19.
J Clin Virol ; 138: 104790, 2021 05.
Article in English | MEDLINE | ID: covidwho-1126919

ABSTRACT

The Roche Cobas SARS-CoV-2 test recently received an Emergency Use Authorization from the U.S. Food and Drug Administration UA for pooling of up to six nasopharyngeal swab samples (NPS). We evaluated the 6-pool approach on both NPS and saliva samples using 564 samples (20 positive NPS and saliva samples each and 262 negative NPS and saliva samples each). The sensitivity of the Roche SARS-CoV-2 RNA test for pooled NPS samples was 100 % (95 %CI: 83.2-100 %) and the sensitivity for pooled saliva samples was 90 % (95 % CI: 68.3-98.8 %). Given the high throughput of the Roche Cobas 6800, pooling of 6 samples has the potential to significantly increase testing capacity without significant loss in sensitivity.


Subject(s)
COVID-19 Nucleic Acid Testing/methods , COVID-19/diagnosis , Nasopharynx/virology , RNA, Viral/isolation & purification , SARS-CoV-2/isolation & purification , Saliva/virology , Specimen Handling , Diagnostic Tests, Routine , Humans , SARS-CoV-2/genetics , Sensitivity and Specificity
20.
Infect Agent Cancer ; 16(1): 17, 2021 Feb 24.
Article in English | MEDLINE | ID: covidwho-1102344

ABSTRACT

Due to the worldwide COVID-19 outbreak it is mandatory for health care workers to develop containment strategies. Recently published data showed, that cancer patients might have a higher risk for severe course of the disease. We therefore developed a strategy of screening and containment for SARS-CoV-2 for hospitalized cancer patients. Our approach includes a temporary isolation in a so-called floating zone and testing strategy for screening of asymptomatic individuals by pooling of samples before RT-PCR amplification. Patients as far as health care professionals got tested twice a week. Nurses and physicians entered the floating zone with full body protection. Within 8 weeks we tested 418 individuals (professionals and patients) in total. Only 2 patients had COVID-19 without documented further transmission of SARS-CoV-2. We therefore think that our strategy might be a useful approach to protect inpatients with cancer at high risk for SARS-CoV-2 infection during this ongoing pandemic.

SELECTION OF CITATIONS
SEARCH DETAIL