Skip to main content
Advertisement
  • Loading metrics

Accuracy of novel antigen rapid diagnostics for SARS-CoV-2: A living systematic review and meta-analysis

Correction

13 Oct 2021: The PLOS Medicine Staff (2021) Correction: Accuracy of novel antigen rapid diagnostics for SARS-CoV-2: A living systematic review and meta-analysis. PLOS Medicine 18(10): e1003825. https://doi.org/10.1371/journal.pmed.1003825 View correction

Abstract

Background

SARS-CoV-2 antigen rapid diagnostic tests (Ag-RDTs) are increasingly being integrated in testing strategies around the world. Studies of the Ag-RDTs have shown variable performance. In this systematic review and meta-analysis, we assessed the clinical accuracy (sensitivity and specificity) of commercially available Ag-RDTs.

Methods and findings

We registered the review on PROSPERO (registration number: CRD42020225140). We systematically searched multiple databases (PubMed, Web of Science Core Collection, medRvix, bioRvix, and FIND) for publications evaluating the accuracy of Ag-RDTs for SARS-CoV-2 up until 30 April 2021. Descriptive analyses of all studies were performed, and when more than 4 studies were available, a random-effects meta-analysis was used to estimate pooled sensitivity and specificity in comparison to reverse transcription polymerase chain reaction (RT-PCR) testing. We assessed heterogeneity by subgroup analyses, and rated study quality and risk of bias using the QUADAS-2 assessment tool. From a total of 14,254 articles, we included 133 analytical and clinical studies resulting in 214 clinical accuracy datasets with 112,323 samples. Across all meta-analyzed samples, the pooled Ag-RDT sensitivity and specificity were 71.2% (95% CI 68.2% to 74.0%) and 98.9% (95% CI 98.6% to 99.1%), respectively. Sensitivity increased to 76.3% (95% CI 73.1% to 79.2%) if analysis was restricted to studies that followed the Ag-RDT manufacturers’ instructions. LumiraDx showed the highest sensitivity, with 88.2% (95% CI 59.0% to 97.5%). Of instrument-free Ag-RDTs, Standard Q nasal performed best, with 80.2% sensitivity (95% CI 70.3% to 87.4%). Across all Ag-RDTs, sensitivity was markedly better on samples with lower RT-PCR cycle threshold (Ct) values, i.e., <20 (96.5%, 95% CI 92.6% to 98.4%) and <25 (95.8%, 95% CI 92.3% to 97.8%), in comparison to those with Ct ≥ 25 (50.7%, 95% CI 35.6% to 65.8%) and ≥30 (20.9%, 95% CI 12.5% to 32.8%). Testing in the first week from symptom onset resulted in substantially higher sensitivity (83.8%, 95% CI 76.3% to 89.2%) compared to testing after 1 week (61.5%, 95% CI 52.2% to 70.0%). The best Ag-RDT sensitivity was found with anterior nasal sampling (75.5%, 95% CI 70.4% to 79.9%), in comparison to other sample types (e.g., nasopharyngeal, 71.6%, 95% CI 68.1% to 74.9%), although CIs were overlapping. Concerns of bias were raised across all datasets, and financial support from the manufacturer was reported in 24.1% of datasets. Our analysis was limited by the included studies’ heterogeneity in design and reporting.

Conclusions

In this study we found that Ag-RDTs detect the vast majority of SARS-CoV-2-infected persons within the first week of symptom onset and those with high viral load. Thus, they can have high utility for diagnostic purposes in the early phase of disease, making them a valuable tool to fight the spread of SARS-CoV-2. Standardization in conduct and reporting of clinical accuracy studies would improve comparability and use of data.

Author summary

Why was this study done?

  • Antigen rapid diagnostic tests (Ag-RDTs) are considered an important diagnostic tool to fight the spread of SARS-CoV-2.
  • An increasing number of Ag-RDTs are offered on the market, and a constantly growing body of literature evaluating their performance is available.
  • To inform decision makers about the best test to choose, an up-to-date summary of their performance is needed.

What did the researchers do and find?

  • On a weekly basis, we search multiple databases for evaluations of Ag-RDTs detecting SARS-CoV-2 and post the results on https://www.diagnosticsglobalhealth.org.
  • Based on the search results up until 30 April 2021, we conducted a systematic review and meta-analysis, including a total of 133 clinical and analytical accuracy studies.
  • Across all meta-analyzed studies, when Ag-RDTs were performed according to manufacturers’ recommendations, they showed a sensitivity of 76.3% (95% CI 73.1% to 79.2%), with LumiraDx (sensitivity 88.2% [95% CI 59.0% to 97.5%]) and, of the instrument-free Ag-RDTs, Standard Q nasal (74.9% sensitivity [95% CI 69.3% to 79.7%]) performing best.
  • Across all Ag-RDTs, sensitivity increased to 95.8% (95% CI 92.3% to 97.8%) when we restricted the analysis to samples with high viral loads (i.e., a Ct value < 25) and to 83.8% (95% CI 76.3% to 89.2%) when tests were performed on patients within the first week after symptom onset.

What do these findings mean?

  • Ag-RDTs detect the vast majority of cases within the first week of symptom onset and those with high viral load. Thus, they can have high utility for diagnostic purposes in the early phase of disease.
  • Out of all assessed tests, LumiraDx showed the highest accuracy. Standard Q was the best-performing test when only considering those that do not require an instrument.
  • A standardization of reporting methods for clinical accuracy studies would enhance future test comparisons.

Introduction

As the COVID-19 pandemic continues around the globe, antigen rapid diagnostic tests (Ag-RDTs) for SARS-CoV-2 are seen as an important diagnostic tool to fight the virus’s spread [1,2]. The number of Ag-RDTs on the market is increasing constantly [3]. Initial data from independent evaluations suggest that the performance of SARS-CoV-2 Ag-RDTs may be lower than what is reported by the manufacturers. In addition, Ag-RDT accuracy seems to vary substantially between tests [46].

With the increased availability of Ag-RDTs, an increasing number of independent validations have been published. Such evaluations differ widely in their quality, methods, and results, making it difficult to assess the true performance of the respective tests [7]. To inform decision makers on the best choice of individual tests, an aggregated, widely available, and frequently updated assessment of the quality, performance, and independence of the data is urgently needed. While other systematic reviews have been published, they include data only up until November 2020 [811], exclude preprints [12], or were industry sponsored [13]. In addition, only 1 assessed the quality of studies in detail, with data up until November 2020 [7,11].

With our systematic review and meta-analysis, we aim to close this gap in the literature and link to a website (https://www.diagnosticsglobalhealth.org) that is regularly updated.

Methods

We developed a study protocol following standard guidelines for systematic reviews [14,15], which is available in S1 Text. We also completed the PRISMA checklist (S1 PRISMA Checklist). Furthermore, we registered the review on PROSPERO (registration number: CRD42020225140).

Search strategy

We performed a search of the databases PubMed, Web of Science, medRxiv, and bioRxiv using search terms that were developed with an experienced medical librarian (M. Grilli) using combinations of subject headings (when applicable) and text-words for the concepts of the search question. The main search terms were “Severe Acute Respiratory Syndrome Corona-virus 2,” “COVID-19,” “Betacoronavirus,” “Coronavirus,” and “Point of Care Testing.” The full list of search terms is available in S2 Text. We also searched the Foundation for Innovative New Diagnostics (FIND) website (https://www.finddx.org/sarscov2-eval-antigen/) for relevant studies manually. We performed the search up until 30 April 2021. No language restrictions were applied.

Inclusion criteria

We included studies evaluating the accuracy of commercially available Ag-RDTs to establish a diagnosis of SARS-CoV-2 infection, against reverse transcription polymerase chain reaction (RT-PCR) or cell culture as reference standard. We included all study populations irrespective of age, presence of symptoms, or study location. We considered cohort studies, nested cohort studies, case–control or cross-sectional studies, and randomized studies. We included both peer-reviewed publications and preprints.

We excluded studies in which patients were tested for the purpose of monitoring or ending quarantine. Also, publications with a population size smaller than 10 were excluded. Although the size threshold of 10 is arbitrary, such small studies are more likely to give unreliable estimates of sensitivity and specificity.

Index tests

Ag-RDTs for SARS-CoV-2 aim to detect infection by recognizing viral proteins. Most Ag-RDTs use specific labeled antibodies attached to a nitrocellulose matrix strip, to capture the virus antigen. Successful binding of the antibodies to the antigen either is detected visually (through the appearance of a line on the matrix strip [lateral flow assay]) or requires a specific reader for fluorescence detection. Microfluidic enzyme-linked immunosorbent assays have also been developed. Ag-RDTs typically provide results within 10 to 30 minutes [6].

Reference standard

Viral culture detects viable virus that is relevant for transmission but is available in research settings only. Since RT-PCR tests are more widely available and SARS-CoV-2 RNA (as reflected by RT-PCR cycle threshold [Ct] value) highly correlates with SARS-CoV-2 antigen quantities, we considered it an acceptable reference standard for the purposes of this systematic review [16]. It is of note that there is currently no international standard for the classification of viral load available.

Study selection and data extraction

Two reviewers (LEB and CE, LEB and SS, or LEB and MB) reviewed the titles and abstracts of all publications identified by the search algorithm independently, followed by a full-text review for those eligible, to select the articles for inclusion in the systematic review. Any disputes were solved by discussion or by a third reviewer (CMD).

A full list of the parameters extracted is included in S1 Table, and the data extraction file is available at https://zenodo.org/record/4924035#.YOlzWS223RZ. Studies that assessed multiple Ag-RDTs or presented results based on differing parameters (e.g., various sample types) were considered as individual datasets.

At first, 4 authors (SK, CE, SS, and MB) extracted 5 randomly selected papers in parallel to align data extraction methods. Afterwards, data extraction and the assessment of methodological quality and independence from test manufacturers (see below) was performed by 1 author per paper (SK, CE, SS, or MB) and controlled by a second (LEB, SK, SS, or MB). Any differences were resolved by discussion or by consulting a third author (CMD).

Study types

We differentiated between clinical accuracy studies (performed on clinical samples) and analytical accuracy studies (performed on spiked samples with a known quantity of virus). Analytical accuracy studies can differ widely in methodology, impeding an aggregation of their results. Thus, while we extracted the data for both kinds of studies, we only considered data from clinical accuracy studies as eligible for the meta-analysis. Separately, we summarized the results of analytical studies and compared them with the results of the meta-analysis for individual tests.

Assessment of methodological quality

The quality of the clinical accuracy studies was assessed by applying the QUADAS-2 tool [17]. The tool evaluates 4 domains: patient selection, index test, reference standard, and flow and timing. For each domain, the risk of bias is analyzed using different signaling questions. Beyond the risk of bias, the tool also evaluates the applicability of each included study to the research question for every domain. The QUADAS-2 tool was adjusted to the needs of this review and can be found in S3 Text.

Assessment of independence from manufacturers

We examined whether a study received financial support from a test manufacturer (including the free provision of Ag-RDTs), whether any study author was affiliated with a test manufacturer, and whether a respective conflict of interest was declared. Studies were judged not to be independent from the test manufacturer if at least 1 of these aspects was present; otherwise, they were considered to be independent.

Statistical analysis and data synthesis

We extracted raw data from the studies and recalculated performance estimates where possible based on the extracted data. The raw data can be found in S2 Table. We prepared forest plots for the sensitivity and specificity of each test and visually evaluated the heterogeneity between studies. If 4 or more datasets were available with at least 20 positive RT-PCR samples per dataset for a predefined analysis, a meta-analysis was performed. We report point estimates of sensitivity and specificity for SARS-CoV-2 detection compared to the reference standard along with 95% confidence intervals (CIs) using a bivariate model (implemented with the “reitsma” command from the R package “mada,” version 0.5.10). When there were fewer than 4 studies for an index test, only a descriptive analysis was performed, and accuracy ranges are reported. In subgroup analyses where papers presented data only on sensitivity, a univariate random-effects inverse variance meta-analysis was performed (using the “metagen” command from the R package “meta,” version 4.11–0). We predefined subgroups for meta-analysis based on the following characteristics: Ct value range, sampling and testing procedure in accordance with manufacturer’s instructions as detailed in the instructions for use (IFU) (henceforth called IFU-conforming) versus not IFU-conforming, age (<18 versus ≥18 years), sample type, presence or absence of symptoms, symptom duration (<7 days versus ≥7 days), viral load, and type of RT-PCR used.

In an effort to use as much of the heterogeneous data as possible, the cutoffs for the Ct value groups were relaxed by 2–3 points within each range. The <20 group included values reported up to ≤20, the <25 group included values reported as ≤24 or <25 or 20–25, and the <30 group included values from ≤29 to ≤33 and 25–30. The ≥25 group included values reported as ≥25 or 25–30, and the ≥30 group included values from ≥30 to ≥35. For the same reason, when categorizing by age, the age group <18 years (children) included samples from persons whose age was reported as <16 or <18 years, whereas the age group ≥18 years (adults) included samples from persons whose age was reported as ≥16 years or ≥18.

For categorization by sample type, we assessed (1) nasopharyngeal (NP) alone or combined with other (e.g., oropharyngeal [OP]), (2) OP alone, (3) anterior nasal (AN) or mid-turbinate (MT), (4) a combination of bronchoalveolar lavage and throat wash (BAL/TW), or (5) saliva. Analyses were preformed using R 4.0.3 (R Foundation for Statistical Computing, Vienna, Austria).

We aimed to do meta-regression to examine the impact of covariates including symptom duration and Ct value range. We also performed the Deeks test for funnel-plot asymmetry as recommended to investigate publication bias for diagnostic test accuracy meta-analyses [18] (using the “midas” command in Stata, version 15); a p-value < 0.10 for the slope coefficient indicates significant asymmetry.

Sensitivity analysis

Two types of sensitivity analyses were planned: estimation of sensitivity and specificity excluding case–control studies, and estimation of sensitivity and specificity excluding non-peer-reviewed studies. We compared the results of each sensitivity analysis against the overall results to assess the potential bias introduced by considering case–control studies and non-peer-reviewed studies.

Results

Summary of studies

The systematic search resulted in 14,254 articles. After removing duplicates, 8,921 articles were screened, and 266 papers were considered eligible for full-text review. Of these, 148 were excluded because they did not present primary data [13,19131] or the Ag-RDT was not commercially available [16,132164], leaving 133 studies to be included in the systematic review (Fig 1) [4,165296].

thumbnail
Fig 1. PRISMA flow diagram.

Based on Page et al. [297]. Ag-RDT, antigen rapid diagnostic test; IFU, instructions for use; sens., sensitivity; spec., specificity.

https://doi.org/10.1371/journal.pmed.1003735.g001

At the end of the data extraction process, 37 studies were still in preprint form [4,171,173,174,177,180,190,192,201,204,205,207,211,214216,218,220,222,223,225,227,231,233,234,238,240,244,247,253,257,265,267,284,287,290,293]. All studies were written in English, except for 2 in Spanish [175,280]. Out of the 133 studies, 9 reported analytical accuracy [173,191,198,208,227,256,274,275,282], and the remaining 124 reported clinical accuracy.

The clinical accuracy studies were divided into 214 datasets, while the 9 analytical accuracy studies accounted for 63 datasets. A total of 61 different Ag-RDTs were evaluated (48 lateral flow with visual readout and 12 requiring an automated reader), with 56 being assessed in a clinical accuracy study. Thirty-nine studies reported data for more than 1 test, and 19 of these studies conducted a head-to-head assessment, i.e., testing at least 2 Ag-RDTs on the same sample or participant. The reference method was RT-PCR in all except 1 study, which used viral culture [281].

The most common reasons for testing were the occurrence of symptoms (55/19.9% of datasets), screening independent of symptoms (19/6.9%), and close contact with a SARS-CoV-2 confirmed case (10/3.6%). In 79 (28.6%) of the datasets, persons were tested due to more than 1 of these reasons, and for 163 datasets (59.1%), the reason for testing was unclear.

In total, 113,242 Ag-RDTs were performed, 112,323 (99.2%) in clinical accuracy studies and 919 (0.8%) in analytical accuracy studies. In the clinical accuracy studies, the mean number of samples per study was 525 (range 16 to 6,954). Only 4,752 (4.2%) tests were performed on pediatric (age group <18 years) samples, and 21,351 (18.9%) on samples from adults (age group ≥18 years). For the remaining 87,139 (76.9%) samples, the age of the persons tested was not specified. Symptomatic patients comprised 36,981 (32.7%) samples; 32,799 (29.0%) samples originated from asymptomatic patients, and for 42,462 (38.4%) samples, the patient’s symptom status could not be identified. The most common sample type evaluated was NP and mixed NP/OP (67,036 samples, 59.2%), followed by AN/MT (27,045 samples, 23.9%). There was substantially less testing done for the other sample types, with 6,254 (5.5%) tests done from OP samples, 1,351 (1.2%) from saliva, and 219 (0.2%) from BAL/TW, and for 11,337 (10.0%) tests, we could not identify the type of sample.

Of the datasets assessing clinical accuracy, 89 (41.6%) involved testing according to the manufacturers’ recommendations (i.e., IFU-conforming), while 100 (46.7%) were not IFU-conforming, and for 25 (11.7%) it was unclear. The most common deviations from the IFU were (1) use of samples that were prediluted in transport media not recommended by the manufacturer (80 datasets; 7 unclear), (2) use of banked samples (60 datasets; 14 unclear), and (3) use of a sample type that was not recommended for the Ag-RDT (17 datasets; 8 unclear).

A summary of the tests evaluated in clinical accuracy studies, including study identification, sample size, sample type, sample condition, and IFU conformity, can be found in Table 1. The Panbio test by Abbott (Germany; henceforth called Panbio) was reported the most frequently, with 39 (18.2%) datasets and 28,089 (25.0%) tests, while the Standard Q test by SD Biosensor (South Korea; distributed in Europe by Roche, Germany; henceforth called Standard Q) was assessed in 37 (17.3%) datasets, with 16,820 (15.0%) tests performed. Detailed results for each clinical accuracy study are available in S1 Fig.

thumbnail
Table 1. Clinical accuracy data for Ag-RDTs against SARS-CoV-2.

https://doi.org/10.1371/journal.pmed.1003735.t001

Methodological quality of studies

The findings on study quality using the QUADAS-2 tool are presented in Figs 2 and 3. In 190 (88.8%) datasets a relevant patient population was assessed. However, for only 44 (20.6%) of the datasets was patient selection considered representative of the setting and population chosen (i.e., they avoided inappropriate exclusions and a case–control design, and enrollment occurred consecutively or randomly).

thumbnail
Fig 2. Methodological quality of the clinical accuracy studies: Risk of bias.

Proportion of studies with low, intermediate, high, or unclear risk of bias (percent).

https://doi.org/10.1371/journal.pmed.1003735.g002

thumbnail
Fig 3. Methodological quality of the clinical accuracy studies: Applicability.

Proportion of studies with low, intermediate, high, or unclear concerns regarding applicability (percent).

https://doi.org/10.1371/journal.pmed.1003735.g003

The conduct and interpretation of the index tests was considered to have low risk for introduction of bias in 113 (52.8%) datasets (through, e.g., appropriate blinding of persons interpreting the visual readout). However, for 99 (46.3%) datasets, sufficient information to clearly judge the risk of bias was not provided. In only 89 (41.6%) datasets were the Ag-RDTs performed according to IFU, while 100 (46.7%) were not IFU-conforming, potentially impacting the diagnostic accuracy (for 25 [11.7%] datasets the IFU status was unclear).

In 81 (37.9%) datasets, the reference standard was performed before the Ag-RDT, or the operator conducting the reference standard was blinded to the Ag-RDT results, resulting in a low risk of bias. In almost all other datasets (132/61.7%), this risk could not be assessed due to missing data. The applicability of the reference test was judged to be of low concern for all datasets, as cell culture and RT-PCR are expected to adequately define the target condition.

In 209 (97.7%) datasets, the sample for the index test and reference test were obtained at the same time, while this was unclear in 5 (2.3%) datasets. All samples included in a dataset were subjected to the same type of RT-PCR in 145 (67.8%) datasets, while different types of RT-PCR were used within the same dataset in 50 (23.4%) datasets. For 19 (8.9%) datasets, it was unclear. Furthermore, for 11 (5.1%) datasets, there was a concern that not all selected patients were included in the analysis.

Finally, 32 (24.1%) of the studies received financial support from the Ag-RDT manufacturer, and in another 9 (6.8%) studies, employment of the authors by the manufacturer of the Ag-RDT studied was indicated. Overall, a competing interest was found in 33 (24.8%) of the studies.

Detection of SARS-CoV-2 infection

Out of 214 clinical datasets (from 124 studies), 20 were excluded from the meta-analysis because they included fewer than 20 RT-PCR positive samples. A further 21 datasets were missing either sensitivity or specificity and were only considered for univariate analyses. Across the remaining 173 datasets, including any test and type of sample, the pooled sensitivity and specificity were 71.2% (95% CI 68.2% to 74.0%) and 98.9% (95% CI 98.6% to 99.1%), respectively. If testing was performed in conformity with IFU, sensitivity increased to 76.3% (95% CI 73.1% to 79.2%), while non-IFU-conforming testing had a sensitivity of 65.9% (95% CI 60.6% to 70.8%). Pooled specificity was similar in both groups (99.1% [95% CI 98.8–99.4%] and 98.3% [95% CI 97.7% to 98.8%], respectively).

Analysis of specific tests

Based on 119 datasets with 71,424 tests performed, we were able to perform bivariate meta-analysis of the sensitivity and specificity for 12 different Ag-RDTs (Fig 4). Across these, the pooled estimates of sensitivity and specificity on all samples were 72.1% (95% CI 68.8% to 75.3%) and 99.0% (95% CI 98.7% to 99.2%), respectively, which were very similar to the overall pooled estimates across all meta-analyzed datasets (71.2% and 98.9%, respectively, above).

thumbnail
Fig 4. Bivariate analysis of 12 antigen rapid diagnostic tests.

Pooled sensitivity and specificity were calculated based on reported sample sizes, true positives, true negatives, false positives, and false negatives.

https://doi.org/10.1371/journal.pmed.1003735.g004

The highest pooled sensitivity was found for the SARS-CoV-2 Antigen Test by LumiraDx (UK; henceforth called LumiraDx) and the Lumipulse G SARS-CoV-2 Ag by Fujirebio (Japan; henceforth called Lumipulse G), with 88.2% (95% CI 59.0% to 97.5%) and 87.2% (95% CI 78.0% to 92.9%), respectively. The Sofia SARS Antigen FIA by Quidel (California, US; henceforth called Sofia) had a pooled sensitivity of 77.4% (95% CI 74.2% to 80.3%). Of the non-instrument tests, the Standard Q and the Standard Q nasal test by SD Biosensor (South Korea; distributed in Europe by Roche, Germany; henceforth called Standard Q nasal) performed best, with a pooled sensitivity of 74.9% (95% CI 69.3% to 79.7%) and 80.2% (95% CI 70.3% to 87.4%), respectively. The pooled sensitivity for Panbio was 71.8% (95% CI 65.4% to 77.5%). Of all Ag-RDTs, the COVID-19 Ag Respi-Strip by Coris BioConcept (Belgium; henceforth called Coris) had the lowest pooled sensitivity, 40.0% (95% CI 28.7% to 52.4%).

The pooled specificity was above 98% for all of the tests, except for the Standard F by SD Biosensor (South Korea) and Lumipulse G, with specificities of 97.7% (95% CI 96.6% to 98.5%) and 96.7% (95% CI 88.6% to 99.1%), respectively. Hierarchical summary receiver operating characteristic values for Standard Q and LumiraDx are available in S2 Fig.

Three Ag-RDTs did not have sufficient data to allow for a bivariate meta-analysis, so a univariate analysis was conducted (Fig 5). For the INNOVA SARS-CoV-2 Antigen Rapid Qualitative Test by Innova Medical Group (California, US), this resulted in a pooled sensitivity and specificity of 76.1% (95% CI 68.1% to 84.1%) and 99.4% (95% CI 98.7% to 100%), respectively. For the NADAL by nal von minden (Germany) and the COVID-19 Rapid Antigen Visual Read by SureScreen Diagnostics (UK), sufficient data were available to analyze only sensitivity, resulting in pooled sensitivity estimates of 58.4% (95% CI 29.2% to 87.6%) and 58.0% (95% CI 38.3% to 77.6%), respectively.

thumbnail
Fig 5. Univariate analysis of 3 antigen rapid diagnostic tests.

Pooled sensitivity and specificity were calculated based on reported sensitivity, specificity, and confidence intervals. SureScreen V, SureScreen Diagnostics COVID-19 Rapid Antigen Visual Read.

https://doi.org/10.1371/journal.pmed.1003735.g005

The remaining 35 Ag-RDTs did not present sufficient data for univariate or bivariate meta-analysis. However, 9/35 had results presented in more than 1 dataset, and these are summarized in Table 2. Herein, the widest ranges of sensitivity were found for the ESPLINE SARS-CoV-2 by Fujirebio (Japan), with sensitivity reported between 8.1% and 80.7%, and the RIDA QUICK SARS-CoV-2 Antigen by R-Biopharm (Germany), with sensitivity between 39.2% and 77.6%, both with 3 datasets each. In contrast, 2 other tests with 2 datasets each showed the least variability in sensitivity: The Zhuhai Encode Medical Engineering SARS-CoV-2 Antigen Rapid Test (China) reported sensitivity between 74.0% and 74.4%, and the COVID-19 Rapid Antigen Fluorescent by SureScreen Diagnostics (UK) reported sensitivity between 60.3% and 69.0%. However, for both tests, both datasets originated from the same studies. Overall, the lowest sensitivity range was reported for the SARS-CoV-2 Antigen Rapid Test by MEDsan (Germany): 36.5% to 45.2% across 2 datasets. The specificity ranges were above 96% for most of the tests. A notable outlier was the 2019-nCov Antigen Rapid Test Kit by Shenzhen Bioeasy Biotechnology (China; henceforth called Bioeasy), reporting the worst, with a specificity as low as 85.6% in 1 study. Forest plots for the datasets for each Ag-RDT are provided in S3 Fig. The remaining 26 Ag-RDTs that were evaluated in 1 dataset only are included in Table 1 S3 Fig.

thumbnail
Table 2. Summary clinical accuracy data for major Ag-RDTs not included in the meta-analysis.

https://doi.org/10.1371/journal.pmed.1003735.t002

In total, 16 studies, accounting for 53 datasets, conducted head-to-head clinical accuracy evaluations of different tests using the same samples from the same participants. These datasets have underlined sample sizes in Table 1; 15 such studies included more than 100 samples, and 1 study included too few samples to draw clear conclusions [286]. Four studies performed their head-to-head evaluation as per manufacturers’ instructions and on symptomatic patients. Across 3 of them, Standard Q (sensitivity 73.2% to 91.2%) and Standard Q nasal (sensitivity 82.5% to 91.2%) showed a similar range of sensitivity [207,215,271]. The fourth reported a sensitivity of 56.4% (95% CI 44.7% to 67.6%) for the Biocredit Covid-19 Ag by RapiGEN (South Korea; henceforth called Rapigen) and 52.6% (95% CI 40.9% to 64.0%) for the SGTi-flex COVID-19 Ag by Sugentech (South Korea) [233].

All other head-to-head comparisons were not IFU-conforming. In one of these, the Rapid COVID-19 Ag Test by Healgen (sensitivity 77.1%) performed better than Standard Q and Panbio (sensitivity 69.8% and 67.7%, respectively) [178]. In contrast to the overall findings of the meta-analysis above, 2 other head-to-head studies found that both Standard Q (sensitivity 43.6% and 49.4%) and Panbio (sensitivity 38.6% and 44.6%) had lower performance than the CLINITEST Rapid COVID-19 Antigen Test by Siemens Healthineers (Germany; henceforth called Clinitest), with reported sensitivity of 51.5% and 54.9% [167,279]. However, another study found both Standard Q and Panbio (sensitivity 81.0% and 82.9%, respectively) to have a higher accuracy than Sofia (sensitivity 80.4%) [196].

Subgroup analyses

The results are presented in Figs 610. Detailed results for the subgroup analyses are available in S4S9 Figs.

thumbnail
Fig 6. Pooled sensitivity by cycle threshold (Ct) values.

Low Ct values are the reverse transcription PCR semi-quantitative correlate for a high virus concentration.

https://doi.org/10.1371/journal.pmed.1003735.g006

thumbnail
Fig 7. Pooled sensitivity and specificity by instructions for use (IFU) conformity.

https://doi.org/10.1371/journal.pmed.1003735.g007

thumbnail
Fig 8. Pooled sensitivity and specificity by sample type.

AN, anterior nasal; MT, mid-turbinate; NP, nasopharyngeal; OP, oropharyngeal.

https://doi.org/10.1371/journal.pmed.1003735.g008

thumbnail
Fig 9. Pooled sensitivity and specificity by presence of symptoms and symptom duration.

https://doi.org/10.1371/journal.pmed.1003735.g009

Subgroup analysis by Ct values.

High sensitivity was achieved for Ct value < 20, at 96.5% (95% CI 92.6% to 98.4%). The pooled sensitivity for Ct value < 25 was markedly better, at 95.8% (95% CI 92.3% to 97.8%), compared to the group with Ct value ≥ 25, at 50.7% (95% CI 35.6% to 65.8%). A similar pattern was observed when the Ct values were analyzed using the cutoffs <30 and ≥30, resulting in a sensitivity of 79.9% (95% CI 70.3% to 86.9%) and 20.9% (95% CI 12.5% to 32.8%), respectively (Fig 6).

In addition, it was possible to meta-analyze test-specific pooled sensitivity for Panbio: 97.7% sensitivity (95% CI 95.3% to 98.9%) for Ct value < 20, 95.8% (95% CI 92.3% to 97.8%) for Ct value < 25, and 83.4% (95% CI 69.1% to 91.9%) for Ct value < 30. Sensitivity was 61.2% (95% CI 38.8% to 79.7%) for Ct value ≥ 25 and 30.5% (95% CI 16.0% to 50.4%) for Ct value ≥ 30. For the other Ag-RDTs only limited data were available, which are presented in S5 Fig.

Subgroup analysis by IFU conformity.

The summary results are presented in Fig 7. When assessing only studies with IFU-conforming testing, pooled sensitivity from 81 datasets with 49,643 samples was 76.3% (95% CI 73.1% to 79.2%). When non-IFU-conforming sampling (75 datasets, 31,416 samples) was performed, sensitivity decreased to 65.9% (95% CI 60.6% to 70.8%).

For 5 tests it was possible to calculate pooled sensitivity estimates including only datasets with IFU-conforming testing: Panbio (sensitivity 76.5% [95% CI 69.5% to 82.3%]; 17 datasets, 12,856 samples), Standard Q (sensitivity 79.3% [95% CI 73.5% to 84.1%]; 15 datasets, 6,584 samples), BinaxNOW (sensitivity 61.8% [95% CI 48.0% to 74.0%]; 4 datasets, 8,163 samples), Rapigen (sensitivity 67.1% [95% CI 50.4% to 80.4%]; 4 datasets, 1,934 samples), and Standard Q nasal (sensitivity 83.8% [95% CI 77.8% to 88.4%]; 5 datasets, 683 samples). Specificity was above 98.6% for all tests.

In contrast, when the Panbio (14 datasets, 9,233 samples) and Standard Q (14 datasets, 4,714 samples) tests were not performed according to IFU, pooled sensitivity decreased to 64.3% (95% CI 50.9% to 75.8%) and 67.4% (95% CI 57.2% to 76.2%), respectively.

Subgroup analysis by sample type.

Most datasets evaluated NP or combined NP/OP swabs (122 datasets and 59,810 samples) as the sample type for the Ag-RDT. NP or combined NP/OP swabs achieved a pooled sensitivity of 71.6% (95% CI 68.1% to 74.9%). Datasets that used AN/MT swabs for Ag-RDTs (32 datasets and 25,814 samples) showed a summary estimate for sensitivity of 75.5% (95% CI 70.4% to 79.9%). This was confirmed by 2 studies that reported direct head-to-head comparison of NP and MT samples from the same participants using the same Ag-RDT (Standard Q), where the 2 sample types showed equivalent performance [271,272]. Analysis of performance with an OP swab (7 datasets, 5,165 samples) showed a pooled sensitivity of only 53.1% (95% CI 40.9% to 65.0%). Saliva swabs (4 datasets, 1,088 samples) showed the lowest pooled sensitivity, at only 37.9% (95% CI 11.8% to 73.5%) (Fig 8).

We were not able to perform a subgroup meta-analysis for BAL/TW due to insufficient data: There was only 1 study with 73 samples evaluating Rapigen, Panbio, and Standard Q [286]. However, BAL/TW would in any case be considered an off-label use.

Subgroup analysis in symptomatic and asymptomatic patients.

Within the datasets possible to meta-analyze, 17,964 (54.1%) samples were from symptomatic, and 15,228 (45.9%) from asymptomatic, patients. The pooled sensitivity for symptomatic patients was markedly different from that of asymptomatic patients: 76.7% (95% CI 70.6% to 81.9%) versus 52.5% (95% CI 43.7% to 61.1%). Specificity was 99% for both groups (Fig 9). Median Ct values differed in symptomatic and asymptomatic patients. For those studies where it was possible to extract a median Ct value, it ranged from 20.5 to 27.0 in symptomatic patients [170,207,226,258,271,272] and from 27.2 to 30.5 in asymptomatic patients [170,201,258].

Subgroup analysis comparing symptom duration.

Data were analyzed for 5,538 patients with symptoms less than 7 days, but very limited data were available for patients with symptoms ≥7 days (397 patients). The pooled sensitivity for patients with onset of symptoms <7 days was 83.8% (95% CI 76.3% to 89.2%), which is markedly higher than the 61.5% (95% CI 52.2% to 70.0%) sensitivity for individuals tested ≥7 days from onset of symptoms (Fig 9).

Subgroup analysis by age.

For adult patients (age ≥ 18 years), it was possible to pool estimates across 3,837 samples, whereas the pediatric group (age < 18 years) included 7,326 samples. Sensitivity and specificity were 64.3% (95% CI 54.7% to 72.9%) and 99.4% (95% CI 98.9% to 99.7%), respectively, in mostly symptomatic patients aged <18 years. In patients aged ≥18 years, sensitivity increased to 74.8% (95% CI 66.5% to 81.6%), while the specificity was similar (98.7%, 95% CI 97.2% to 99.4%) (Fig 10).

Subgroup analysis by type of RT-PCR and viral load.

We were not able to perform a meta-analysis for the subgroups by type of RT-PCR or viral load (viral copies/mL) due to insufficient data.

In 152 (71.0%) of the datasets only 1 type of RT-PCR was used, whereas 37 (17.3%) of the datasets tested samples in the same dataset using different RT-PCR methods. For 25 (11.7%) of the datasets, the type of RT-PCR was not reported. The Cobas SARS-CoV-2 Test from Roche (Germany) was used most frequently, in 63 (29.4%) of the datasets, followed by the Allplex 2019-nCoV Assay from Seegene in 41 (19.2%) and the SARS-CoV-2 assay from TaqPath in 20 (9.3%) of the datasets.

Median sensitivity was 72.4% (range 46.9% to 100%) in samples with viral load > 5 log10 copies/mL, 97.8% (range 71.4% to 100%) for >6 log10 copies/mL, and 100% (range 93.8% to 100%) for >7 log10 copies/mL, showing that the sensitivity increases with increasing viral load.

Meta regression.

We were not able to perform a meta-regression due to the considerable heterogeneity in reporting subgroups, which resulted in too few studies with sufficient data for comparison.

Publication bias.

The result of the Deeks test (p = 0.001) shows significant asymmetry in the funnel plot for all datasets with complete results. This indicates there may be publication bias from studies with small sample sizes. The funnel plot is presented in S10 Fig.

Comparison with analytical studies

The 9 analytical studies were divided into 63 datasets, evaluating 23 different Ag-RDTs. Only 7 studies reported a sample size, for which 833 (90.6%) samples originated from NP swabs, while for 86 (9.4%) the sample type was unclear. One of the 2 studies not reporting sample size used saliva samples [198], while the other used the sample type specified in the respective Ag-RDT’s IFU [173].

Overall, the reported analytical sensitivity (limit of detection [LOD]) in the studies resembled the results of the meta-analysis presented above. Rapigen (LOD, in log10 copies per swab: 10.2) and Coris (LOD 7.46) were found to perform worse than Panbio (LOD 6.6 to 6.1) and Standard Q (LOD 6.8 to 6.0), whereas Clinitest (LOD 6.0) and BinaxNOW by Abbott (LOD 4.6 to 4.9) performed better [191,256,282]. Similar results were found in another study, where Standard Q showed the lowest LOD (detecting virus up to what is an equivalent Ct value of 26.3 to 28.7), compared to that of Rapigen and Coris (detecting virus up to what is an equivalent Ct value of only 18.4 for both) [208,274,275]. However, another study found Panbio, Standard Q, Coris, and BinaxNOW to have a similar LOD values of 5.0 × 103 plaque forming units (PFU)/mL, but the ESPLINE SARS-CoV-2 by Fujirebio (Japan), the COVID-19 Rapid Antigen Test by Mologic (UK), and the Sure Status COVID-19 Antigen Card Test by Premier Medical Corporation (India) performed markedly better (LOD 2.5 × 102 to 5.0 × 102 PFU/mL) [173]. An overview of all LOD values reported in the studies can be found in S3 Table.

Sensitivity analysis

When the datasets from case–control studies (25/173) were excluded, the estimated sensitivity did not differ greatly, with a value of 70.9% (95% CI 67.7% to 73.9%), compared to 71.2% (95% CI 68.2% to 74.0%) in the overall analysis, with no change in pooled specificity. When the datasets from preprints (64/173) were excluded, sensitivity decreased slightly, to 67.2% (95% CI 62.9% to 71.3%), compared to the overall analysis.

Discussion

In this comprehensive systematic review and meta-analysis, we have summarized the data of 133 studies evaluating the accuracy of 61 different Ag-RDTs. Across all meta-analyzed samples, our results show a pooled sensitivity and specificity of 71.2% (95% CI 68.2% to 74.0%) and 98.9% (95% CI 98.6% to 99.1%), respectively. Over half of the studies did not perform the Ag-RDT in accordance with the test manufacturers’ recommendation, or the performance was unknown, which negatively impacted the sensitivity. When we considered only IFU-conforming studies, the sensitivity increased to 76.3% (95% CI 73.1% to 79.2%). While we found the sensitivity to vary across specific tests, the specificity was consistently high.

The 2 Ag-RDTs that have been approved through the WHO emergency use listing procedure, Abbott Panbio and SD Biosensor Standard Q (distributed by Roche in Europe), have not only drawn the largest research interest, but also perform at or above average when their pooled accuracy is compared to that of all Ag-RDTs (sensitivity of 71.8% for Panbio and 74.9% for Standard Q). Standard Q nasal demonstrated an even higher pooled sensitivity (80.2% compared to the NP test), although this is likely due to variability in the populations tested, as head-to-head performance showed a comparable sensitivity. Three other Ag-RDTs showed an even higher accuracy, with sensitivities ranging from 77.4% to 88.2% (namely Sofia, Lumipulse G, and LumiraDx), but were only assessed on relatively small samples sizes (ranging from 1,373 to 3,532), and all required an instrument/reader.

Not surprisingly, lower Ct values, the RT-PCR semi-quantitative correlate for high virus concentration, resulted in significantly higher Ag-RDT sensitivity than higher Ct values (pooled sensitivity 96.5% and 95.8% for Ct value < 20 and <25, respectively, versus 50.7% and 20.9% for Ct value ≥ 25 and ≥30, respectively). This confirms prior data that suggested that antigen concentrations and Ct values were highly correlated in NP samples [16]. Ag-RDTs also showed higher sensitivity in patients within 7 days after symptom onset compared to patients later in the course of the disease (pooled sensitivity 83.8% versus 61.5%), which is to be expected given that samples from patients within the first week after symptom onset have been shown to contain the highest virus concentrations [298]. In line with this, studies reporting an unexpectedly low overall sensitivity either shared a small population size with an on average high Ct value [230,273,288] or performed the Ag-RDT not as per IFU, e.g., using saliva or prediluted samples [167,170,203,248,279]. In contrast, studies with an unusually high Ag-RDT sensitivity were based on study populations with a low median Ct value, between 18 and 22 [189,255,284].

Our analysis also found that the accuracy of Ag-RDTs is substantially higher in symptomatic patients than in asymptomatic patients (pooled sensitivity 76.7% versus 52.5%). This is not surprising as studies that enrolled symptomatic patients showed a lower range of median Ct values (i.e., higher viral load) than studies enrolling asymptomatic patients. Given that other studies found symptomatic and asymptomatic patients to have comparable viral loads [299,300], the differences found in our analysis are likely explained by the varied time in the course of the disease at which testing is performed in asymptomatic patients presenting for one-time screening testing. Because symptoms start in the early phase of the disease, when viral load is still high, studies testing only symptomatic patients have a higher chance of including patients with high viral loads. In contrast, study populations drawn from only asymptomatic patients have a higher chance of including patients at any point of disease (i.e., including late in disease, when PCR is still positive, but viable virus is rapidly decreasing) [301].

With regards to the sampling and testing procedure, we found Ag-RDTs to perform similarly across upper respiratory swab samples (e.g., NP and AN/MT), particularly when considering the most reliable comparisons from head-to-head studies.

Similar to previous assessment [7], the methodological quality of the included studies revealed a very heterogenous picture. In the future, aligning the design of clinical accuracy studies with common agreed-upon minimal specifications (e.g., by WHO or the European Centre for Disease Control and Prevention) and reporting the results in a standardized way [302] would improve data quality and comparability.

The main strengths of our study lie in its comprehensive approach and continuous updates. By linking this review to our website, https://www.diagnosticsglobalhealth.org, we strive to equip decision makers with the latest research findings on Ag-RDTs for SARS-CoV-2 and, to the best of our knowledge, are the first in doing so. At least once per week the website is updated by continuing the literature search and process described above. We plan to update the meta-analysis on a monthly basis and publish it on the website. Furthermore, our study used rigorous methods as both the study selection and data extraction were performed by one author and independently validated by a second, we conducted blinded pilot extractions before of the actual data extraction, and we prepared a detailed interpretation guide for the QUADAS-2 tool.

The study may be limited by the inclusion of both preprints and peer-reviewed literature, which could affect the quality of the data extracted. However, we aimed to balance this potential effect by applying a thorough assessment of all clinical studies included, utilizing the QUADAS-2 tool, and performing a sensitivity analysis excluding preprint manuscripts. In addition, the studies included in our analysis varied widely in the reported range of viral loads, limiting the comparability of their results. To control for this, we analyzed the Ag-RDTs’ performance at different levels of viral load. Finally, even though we are aware that further data exist from other sources, for example from governmental research institutes [303], such data could not be included because sufficiently detailed descriptions of the methods and results are not publicly available.

Conclusion

In summary, it can be concluded that there are Ag-RDTs available that have high sensitivity for the detection of a SARS-CoV-2 infection—particularly when performed in the first week of illness, when viral load is high—and excellent specificity. However, our analysis also highlights the variability in results between tests (which is not reflected in the manufacturer-reported data), indicating the need for independent validations. Furthermore, the analysis highlights the importance of performing tests in accordance with the manufacturers’ recommended procedures, and in alignment with standard diagnostic evaluation and reporting guidelines. The accuracy achievable by the best-performing Ag-RDTs, combined with the rapid turnaround time compared to RT-PCR, suggests that these tests could have a significant impact on the pandemic if applied in thoughtful testing and screening strategies.

Supporting information

S1 Fig. Detailed results of the QUADAS-2 assessment.

https://doi.org/10.1371/journal.pmed.1003735.s001

(PDF)

S2 Fig. Hierarchical summary receiver operating characteristic curve for Standard Q Ag-RDT.

https://doi.org/10.1371/journal.pmed.1003735.s002

(PDF)

S4 Fig. Forest plots for subgroup analysis by Ct value.

https://doi.org/10.1371/journal.pmed.1003735.s004

(PDF)

S5 Fig. Forest plots for subgroup analysis by Ct value per test.

https://doi.org/10.1371/journal.pmed.1003735.s005

(PDF)

S6 Fig. Forest plots for subgroup analysis by IFU versus non-IFU.

https://doi.org/10.1371/journal.pmed.1003735.s006

(PDF)

S7 Fig. Forest plots for subgroup analysis by sample type.

https://doi.org/10.1371/journal.pmed.1003735.s007

(PDF)

S8 Fig. Forest plots for subgroup analysis by symptomatic versus asymptomatic.

https://doi.org/10.1371/journal.pmed.1003735.s008

(PDF)

S9 Fig. Forest plots for subgroup analysis by symptom duration.

https://doi.org/10.1371/journal.pmed.1003735.s009

(PDF)

S10 Fig. Funnel plot test for all datasets included in the meta-analysis.

https://doi.org/10.1371/journal.pmed.1003735.s010

(PDF)

S1 Table. List of data items extracted from studies.

https://doi.org/10.1371/journal.pmed.1003735.s012

(XLSX)

S1 Text. Study protocol submitted to PROSPERO (registration: CRD42020225140).

https://doi.org/10.1371/journal.pmed.1003735.s015

(DOCX)

S3 Text. QUADAS-2 assessment interpretation guide.

https://doi.org/10.1371/journal.pmed.1003735.s017

(DOCX)

References

  1. 1. World Health Organization. Antigen-detection in the diagnosis of SARS-CoV-2 infection using rapid immunoassays: interim guidance, 11 September 2020. WHO/2019-nCoV/Antigen_Detection/2020.1. Geneva: World Health Organization; 2020.
  2. 2. Mina MJ, Parker R, Larremore DB. Rethinking Covid-19 test sensitivity—a strategy for containment. N Engl J Med. 2020;383(22):e120. pmid:32997903
  3. 3. Federal Institute for Drugs and Medical Devices. Antigen-Tests auf SARS-CoV-2. Bonn: Federal Institute for Drugs and Medical Devices; 2021 [cited 2021 Jun 7]. https://www.bfarm.de/DE/Medizinprodukte/Antigentests/_node.html.
  4. 4. Krüger LJ, Gaeddert M, Köppel L, Brümmer LE, Gottschalk C, Miranda IB, et al. Evaluation of the accuracy, ease of use and limit of detection of novel, rapid, antigen-detecting point-of-care diagnostics for SARS-CoV-2. medRxiv. 2020 Oct 4.
  5. 5. Denkinger CM, Grenier J, Minion J, Pai M. Promise versus reality: optimism bias in package inserts for tuberculosis diagnostics. J Clin Microbiol. 2012;50(7):2455–61. pmid:22573592
  6. 6. World Health Organization. Advice on the use of point-of-care immunodiagnostic tests for COVID-19: scientific brief, 8 April 2020. WHO/2019-nCoV/Sci_Brief/POC_immunodiagnostics/2020.1. Geneva: World Health Organization; 2020.
  7. 7. Dinnes J, Deeks JJ, Adriano A, Berhane S, Davenport C, Dittrich S, et al. Rapid, point-of-care antigen and molecular-based tests for diagnosis of SARS-CoV-2 infection. Cochrane Database Syst Rev. 2020;8(8):CD013705. pmid:32845525
  8. 8. Olalekan A, Iwalokun B, Akinloye OM, Popoola O, Samuel TA, Akinloye O. COVID-19 rapid diagnostic test could contain transmission in low- and middle-income countries. Afr J Lab Med. 2020;9(1):1255. pmid:33102170
  9. 9. Castro R, Luz PM, Wakimoto MD, Veloso VG, Grinsztejn B, Perazzo H. COVID-19: a meta-analysis of diagnostic test accuracy of commercial assays registered in Brazil. Braz J Infect Dis. 2020;24(2):180–7. pmid:32330437
  10. 10. La Marca A, Capuzzo M, Paglia T, Roli L, Trenti T, Nelson SM. Testing for SARS-CoV-2 (COVID-19): a systematic review and clinical guide to molecular and serological in-vitro diagnostic assays. Reprod Biomed Online. 2020;41(3):483–99. pmid:32651106
  11. 11. Dinnes J, Deeks JJ, Berhane S, Taylor M, Adriano A, Davenport C, et al. Rapid, point-of-care antigen and molecular-based tests for diagnosis of SARS-CoV-2 infection. Cochrane Database Syst Rev. 2021;3:CD013705. pmid:33760236
  12. 12. Van Walle I, Leitmeyer K, Broberg E. Meta-analysis of the clinical performance of commercial SARS-CoV-2 nucleic acid, antigen and antibody tests up to 22 August 2020. medRxiv. 2020 Sep 18.
  13. 13. Hayer J, Kasapic D, Zemmrich C. Real-world clinical performance of commercial SARS-CoV-2 rapid antigen tests in suspected COVID-19: a systematic meta-analysis of available data as per November 20, 2020. medRxiv. 2020 Dec 4.
  14. 14. Leeflang MM, Deeks JJ, Gatsonis C, Bossuyt PM, Cochrane Diagnostic Test Accuracy Working Goup. Systematic reviews of diagnostic test accuracy. Ann Intern Med. 2008;149(12):889–97. pmid:19075208
  15. 15. Moher D, Liberati A, Tetzlaff J, Altman DG, PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann Intern Med. 2009;151(4):264–9, W64. pmid:19622511
  16. 16. Pollock N, Savage T, Wardell H, Lee R, Mathew A, Stengelin M, et al. Correlation of SARS-CoV-2 nucleocapsid antigen and RNA concentrations in nasopharyngeal samples from children and adults using an ultrasensitive and quantitative antigen assay. medRxiv. 2020 Nov 13.
  17. 17. Whiting PF, Rutjes AW, Westwood ME, Mallett S, Deeks JJ, Reitsma JB, et al. QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med. 2011;155(8):529–36. pmid:22007046
  18. 18. van Enst WA, Ochodo E, Scholten RJ, Hooft L, Leeflang MM. Investigation of publication bias in meta-analyses of diagnostic test accuracy: a meta-epidemiological study. BMC Med Res Methodol. 2014;14:70. pmid:24884381
  19. 19. Courtellemont L, Guinard J, Guillaume C, Giaché S, Rzepecki V, Seve A, et al. Real-life performance of a novel antigen detection test on nasopharyngeal specimens for SARS-CoV-2 infection diagnosis: a prospective study. medRxiv. 2020 Nov 3.
  20. 20. Diao B, Wen K, Chen J, Liu Y, Yuan Z, Han C, et al. Diagnosis of acute respiratory syndrome coronavirus 2 infection by detection of nucleocapsid protein. medRxiv. 2020 Mar 13. pmid:33031947
  21. 21. Foundation for Innovative New Diagnostics. FIND evaluation of Abbott Panbio COVID-19 Ag Rapid Test Device. External Report Version 21, 10 December 2020. Geneva: Foundation for Innovative New Diagnostics; 2020.
  22. 22. Foundation for Innovative New Diagnostics. FIND evaluation of Coris BioConcept COVID-19 Ag Respi-Strip. External Report Version 12, 10 December 2020. Geneva: Foundation for Innovative New Diagnostics; 2020.
  23. 23. Foundation for Innovative New Diagnostics. FIND evaluation of Shenzhen Bioeasy Biotechnology Co. Ltd. 2019-nCoV Ag Rapid Test Kit (Fluorescence). External Report Version 10, 11 February 2021. Geneva: Foundation for Innovative New Diagnostics; 2021.
  24. 24. Ikeda M, Imai K, Tabata S, Miyoshi K, Mizuno T, Murahara N, et al. Clinical evaluation of self-collected saliva by RT-qPCR, direct RT-qPCR, RT-LAMP, and a rapid antigen test to diagnose COVID-19. medRxiv. 2020 Jun 8.
  25. 25. Klein JAF, Krüger L, Tobian F, Gaeddert M, Lainati F, Schnitzler P, et al. Head-to-head performance comparison of self-collected nasal versus professional-collected nasopharyngeal swab for a WHO-listed SARS-CoV-2 antigen-detecting rapid diagnostic test. medRxiv. 2021 Mar 24. pmid:34028625
  26. 26. Masiá M, Fernández-González M, Sánchez M, Carvajal M, García JA, Gonzalo N, et al. Nasopharyngeal Panbio COVID-19 antigen performed at point-of-care has a high sensitivity in symptomatic and asymptomatic patients with higher risk for transmission and older age. medRxiv. 2020 Nov 17.
  27. 27. Weitzel T, Legarraga P, Iruretagoyena M, Pizarro G, Vollrath V, Araos R, et al. Head-to-head comparison of four antigen-based rapid detection tests for the diagnosis of SARS-CoV-2 in respiratory samples. bioRxiv. 2020 May 30.
  28. 28. Peeling RW, Olliaro P. Rolling out COVID-19 antigen rapid diagnostic tests: the time is now. Lancet Infect Dis. 2021;21(8):1052–3. pmid:33773617
  29. 29. Cui Z, Chang H, Wang H, Lim B, Hsu CC, Yu Y, et al. Development of a rapid test kit for SARS-CoV-2: an example of product design. Biodes Manuf. 2020 May 11. pmid:32395391
  30. 30. Ndwandwe D, Mathebula L, Kamadjeu R, Wiysonge CS. Cochrane corner: rapid point-of-care antigen and molecular-based tests for the diagnosis of COVID-19 infection. Pan Afr Med J. 2020;37(Suppl 1):10. pmid:33294111
  31. 31. Pavelka M, Van-Zandvoort K, Abbott S, Sherratt K, Majdan M, Jarčuška P, et al. The effectiveness of population-wide, rapid antigen test based screening in reducing SARS-CoV-2 infection prevalence in Slovakia. medRxiv. 2020 Dec 4.
  32. 32. Ebrahimi M, Harmooshi NN, Rahim F. Diagnostic utility of antigen detection rapid diagnostic tests for Covid- 19: a systematic review and meta-analysis. medRxiv. 2021 Apr 5.
  33. 33. Everitt ML, Tillery A, David MG, Singh N, Borison A, White IM. A critical review of point-of-care diagnostic technologies to combat viral pandemics. Anal Chim Acta. 2021;1146:184–99. pmid:33461715
  34. 34. Hledík M, Polechová J, Beiglböck M, Herdina AN, Strassl R, Posch M. Analysis of the specificity of the SD Biosensor Standard Q Ag-Test based on Slovak mass testing data. medRxiv. 2020 Dec 11.
  35. 35. Huergo MAC, Thanh NTK. Current advances in the detection of COVID-19 and evaluation of the humoral response. Analyst. 2021;146:382–402. pmid:33410826
  36. 36. Laghrib F, Saqrane S, El Bouabi Y, Farahi A, Bakasse M, Lahrich S, et al. Current progress on COVID-19 related to biosensing technologies: new opportunity for detection and monitoring of viruses. Microchem J. 2021;160:105606. pmid:33052148
  37. 37. Maddali H, Miles CE, Kohn J, O’Carroll DM. Optical biosensors for virus detection: prospects for SARS-CoV-2/COVID-19. Chembiochem. 2021;22(7):1176–89. pmid:33119960
  38. 38. Marchán-López Á, García BA. Diagnostic performance of antigen testing for SARS-CoV-2. J Pediatr. 2021;233:283. pmid:33631167
  39. 39. McDermott JH, Stoddard D, Ellingford JM, Gokhale D, Reynard C, Black G, et al. Utilizing point-of-care diagnostics to minimize nosocomial infection in the 2019 novel coronavirus (SARS-CoV-2) pandemic. QJM. 2020;113(12):851–3. pmid:32492142
  40. 40. Moreira V, Mascarenhas P, Machado V, Botelho J, Mendes JJ, Taveira N, et al. Diagnosis of SARS-Cov-2 infection using specimens other than naso- and oropharyngeal swabs: a systematic review and meta-analysis. Diagnostics. 2021;11:363. pmid:33670020
  41. 41. Nimmo C, Agbetile J, Bhowmik A, Capocci S, Rajakulasingam RK. Implementing rapid diagnostics for COVID-19. Lancet Respir Med. 2021;9(1):e7. pmid:33189159
  42. 42. Raimann FJ, Piekarski F, Adam EH, Zacharowski K, Neef V. Safety considerations for the use of Point-Of-Care diagnostics during SARS-CoV-2 pandemic. J Clin Lab Anal. 2021;35(1):e23631. pmid:33089548
  43. 43. Razmy AM, Junaideen SM. Issues of random sampling with rapid antigen tests for COVID-19 diagnosis: a special reference to Kalmunai RDHS Division. medRxiv. 2021 Jan 20.
  44. 44. Salcedo N, Harmon A, Herrera BB. Pooling of samples for SARS-CoV-2 detection using rapid antigen tests. medRxiv. 2021 Feb 12.
  45. 45. Sanderlin JS, Golding JD, Wilcox T, Mason DH, McKelvey KS, Pearson DE, et al. Occupancy modeling and resampling overcomes low test sensitivity to produce accurate SARS-CoV-2 prevalence estimates. BMC Public Health. 2021;21(1):577. pmid:33757468
  46. 46. Scheier T, Schibli A, Eich G, Rüegg C, Kube F, Schmid A, et al. Universal admission screening for SARS-CoV-2 infections among hospitalized patients, Switzerland, 2020. Emerg Infect Dis. 2021;27(2):404–10. pmid:33395382
  47. 47. Stovitz SD. In suspected SARS-CoV-2, rapid antigen detection tests had 67% to 73% sensitivity and 98% to 100% specificity. Ann Intern Med. 2021;174:JC56. pmid:33939481
  48. 48. van Beek J, Igloi Z, Boelsums T, Fanoy E, Gotz H, Molenkamp R, et al. From more testing to smart testing: data-guided SARS-CoV-2 testing choices. medRxiv. 2020 Oct 14.
  49. 49. Weiss G, Bellmann-Weiler R. Rapid antigen testing and non-infectious shedding of SARS-Cov2. Infection. 2021;49(4):789–90. pmid:33386584
  50. 50. Ehrenberg A, Moehle E, Brook C, Doudna Cate A, Witkowsky L, Sachdeva R, et al. Launching a saliva-based SARS-CoV-2 surveillance testing program on a university campus. medRxiv. 2021 Jan 30. pmid:34038425
  51. 51. Boďová K, Kollár R. Characteristic spatial scales of SARS-CoV-2 pandemics: lessons from mass rapid antigen testing in Slovakia. medRxiv. 2020 Dec 24.
  52. 52. Canadian Public Health Laboratory Network. Interim guidance on the use of the Abbott Panbio COVID-19 Antigen Rapid Test. Can Commun Dis Rep. 2021;47(1):17–22. pmid:33767600
  53. 53. Crozier A, Rajan S, Buchan I, McKee M. Put to the test: use of rapid testing technologies for covid-19. BMJ. 2021;372:n208. pmid:33536228
  54. 54. D’Agostino McGowan L, Lee E, Grantz K, Kucirka L, Gurley E, Lessler J. Testing out of quarantine. medRxiv. 2021 Feb 1.
  55. 55. Rapid and frequent testing. Nat Biomed Eng. 2020;4(12):1121–2. pmid:33293723
  56. 56. Fitzpatrick MC, Pandey A, Wells CR, Sah P, Galvani AP. Buyer beware: inflated claims of sensitivity for rapid COVID-19 tests. Lancet. 2021;397(10268):24–5. pmid:33333002
  57. 57. Frnda J, Durica M. On pilot massive COVID-19 testing by antigen tests in Europe. Case study: Slovakia. Infect Dis Rep. 2021;13(1):45–57. pmid:33430283
  58. 58. Ghaffari A, Meurant R, Ardakani A. COVID-19 point-of-care diagnostics that satisfy global target product profiles. Diagnostics. 2021;11(1):115. pmid:33445727
  59. 59. Ruhan A, Wang H, Wang W, Tan W. Summary of the detection kits for SARS-CoV-2 approved by the National Medical Products Administration of China and their application for diagnosis of COVID-19. Virol Sin. 2020;35:699–712. pmid:33351166
  60. 60. Abdelrazik AM, Elshafie SM, Abdelaziz HM. Potential use of antigen-based rapid test for SARS-CoV-2 in respiratory specimens in low-resource settings in Egypt for symptomatic patients and high-risk contacts. Lab Med. 2020;52:e46–9. pmid:33283230
  61. 61. Aoki K, Nagasawa T, Ishii Y, Yagi S, Okuma S, Kashiwagi K, et al. Clinical validation of quantitative SARS-CoV-2 antigen assays to estimate SARS-CoV-2 viral loads in nasopharyngeal swabs. J Infect Chemother. 2020;27(4):613–6. pmid:33423918
  62. 62. Bonde J, Ejegod D, Pedersen H, Smith B, Cortes D, Leding C, et al. Clinical validation of point-of-care SARS-COV-2 BD Veritor antigen test by a single throat swab for rapid COVID-19 status on hospital patients predominantly without overt COVID symptoms. medRxiv. 2021 Apr 17.
  63. 63. Boum Y, Fai KN, Nicolay B, Mboringong AB, Bebell LM, Ndifon M, et al. Performance and operational feasibility of antigen and antibody rapid diagnostic tests for COVID-19 in symptomatic and asymptomatic patients in Cameroon: a clinical, prospective, diagnostic accuracy study. Lancet Infect Dis. 2021;21(8):1089–96. pmid:33773618
  64. 64. Courtellemont L, Guinard J, Guillaume C, Giaché S, Rzepecki V, Seve A, et al. High performance of a novel antigen detection test on nasopharyngeal specimens for diagnosing SARS-CoV-2 infection. J Med Virol. 2021;93(5):3152–7. pmid:33615487
  65. 65. Eshghifar N, Busheri A, Shrestha R, Beqaj S. Evaluation of analytical performance of seven rapid antigen detection kits for detection of SARS-CoV-2 virus. Int J Gen Med. 2021;14:435–40. pmid:33603450
  66. 66. Hingrat QL, Visseaux B, Laouenan C, Tubiana S, Bouadma L, Yazdanpanah Y, et al. Detection of SARS-CoV-2 N-antigen in blood during acute COVID-19 provides a sensitive new marker and new testing alternatives. Clin Microbiol Infect. 2020;27(5):789.e1–5. pmid:33307227
  67. 67. Kobayashi R, Murai R, Asanuma K, Fujiya Y, Takahashi S. Evaluating a novel, highly sensitive, and quantitative reagent for detecting SARS-CoV-2 antigen. J Infect Chemother. 2021;27(6):800–7. pmid:33546959
  68. 68. Kritikos A, Caruana G, Brouillet R, Miroz J-P, Samia A-M, Geraldine S, et al. Sensitivity of rapid antigen testing and RT-PCR performed on nasopharyngeal swabs versus saliva samples in COVID-19 hospitalized patients: results of a prospective comparative trial (RESTART). medRxiv. 2021 Apr 15.
  69. 69. Lanser L, Bellmann-Weiler R, Öttl KW, Huber L, Griesmacher A, Theurl I, et al. Evaluating the clinical utility and sensitivity of SARS-CoV-2 antigen testing in relation to RT-PCR Ct values. Infection. 2021;49(3):555–7. pmid:33185807
  70. 70. Lv Y, Ma Y, Si Y, Zhu X, Zhang L, Feng H, et al. Rapid SARS-CoV-2 antigen detection potentiates early diagnosis of COVID-19 disease. Biosci Trends. 2021;15(2):93–9. pmid:33776018
  71. 71. Miyakawa K, Funabashi R, Yamaoka Y, Jeremiah SS, Katada J, Wada A, et al. SARS-CoV-2 antigen rapid diagnostic test enhanced with silver amplification technology. medRxiv. 2021 Jan 31.
  72. 72. Nagura-Ikeda M, Imai K, Tabata S, Miyoshi K, Murahara N, Mizuno T, et al. Clinical evaluation of self-collected saliva by quantitative reverse transcription-PCR (RT-qPCR), direct RT-qPCR, reverse transcription-loop-mediated isothermal amplification, and a rapid antigen test to diagnose COVID-19. J Clin Microbiol. 2020;58(9):e01438–20. pmid:32636214
  73. 73. Oh SM, Jeong H, Chang E, Choe PG, Kang CK, Park WB, et al. Clinical application of the standard Q COVID-19 Ag Test for the detection of SARS-CoV-2 infection. J Korean Med Sci. 2021;36(14):e101. pmid:33847084
  74. 74. Peña-Rodrígez M, Viera-Segura O, García-Chagollán M, Zepeda-Nuño JS, Muñoz-Valle JF, Mora-Mora J, et al. Performance evaluation of a lateral flow assays for nasopharyngeal antigen detection for SARS-CoV-2 diagnosis. J Clin Lab Anal. 2021;35:e23745. pmid:33675086
  75. 75. Regev-Yochay G, Kriger O, Beni S, Rubin C, Mina M, Mechnik B, et al. Real world performance of SARS-CoV-2 antigen rapid diagnostic tests in various clinical settings. medRxiv. 2021 Mar 5.
  76. 76. Ren A, Sohaei D, Zacharioudakis I, Sigal G, Stengelin M, Matthew A, et al. Ultrasensitive assay for saliva-based SARS-CoV-2 antigen detection. medRxiv. 2021 Feb 19.
  77. 77. Rodrigues J, Gouveia C, Santos MA, Costa O, Côrte-Real R, Brito MJ. Comparison of nasopharyngeal samples for SARS-CoV-2 detection in a paediatric cohort. J Paediatr Child Health. 2021;57(7):1078–81. pmid:33605504
  78. 78. Saeed U, Uppal SR, Piracha ZZ, Rasheed A, Aftab Z, Zaheer H, et al. Evaluation of SARS-CoV-2 antigen-based rapid diagnostic kits in Pakistan: formulation of COVID-19 national testing strategy. Virol J. 2021;18(1):34. pmid:33581714
  79. 79. Smith R, Gibson L, Martinez P, Ke R, Mirza A, Conte M, et al. Longitudinal assessment of diagnostic test performance over the course of acute SARS-CoV-2 infection. medRxiv. 2021 Mar 22. pmid:33791719
  80. 80. Stokes W, Berenger B, Portnoy D, Scott B, Szelewicki J, Singh T, et al. Real-world clinical performance of the Abbott Panbio with nasopharyngeal, throat and saliva swabs among symptomatic individuals with COVID-19. medRxiv. 2021 Jan 21.
  81. 81. Thommes L, Burkert FR, Öttl KW, Goldin D, Loacker L, Lanser L, et al. Comparative evaluation of four SARS-CoV-2 antigen tests in hospitalized patients. Int J Infect Dis. 2021;105:144–6. pmid:33609774
  82. 82. Yokota I, Sakurazawa T, Sugita J, Iwasaki S, Yasuda K, Yamashita N, et al. Performance of qualitative and quantitative antigen tests for SARS-CoV-2 in early symptomatic patients using saliva. medRxiv. 2020 Nov 10.
  83. 83. Zacharias M, Stangl V, Thüringer A, Loibner M, Wurm P, Wolfgruber S, et al. Rapid antigen test for postmortem evaluation of SARS-CoV-2 carriage. Emerg Infect Dis. 2021;27(6):1734–7. pmid:33848454
  84. 84. Apostolou T, Kyritsi M, Vontas A, Loizou K, Hadjilouka A, Speletas M, et al. Development and performance characteristics evaluation of a new Bioelectric Recognition Assay (BERA) method for rapid Sars-CoV-2 detection in clinical samples. J Virol Methods. 2021;293:114166. pmid:33872651
  85. 85. Arnaout R, Lee RA, Lee GR, Callahan C, Cheng A, Yen CF, et al. The limit of detection matters: the case for benchmarking severe acute respiratory syndrome coronavirus 2 testing. Clin Infect Dis. 2021 Feb 3. pmid:33532847
  86. 86. Cheah PK, Ongkili DF, Zaharuddin FS, Hashim MI, Ho CV, Lee HG, et al. Discrepancy in screening performances of different rapid test kits for SARS-CoV-2; a letter to editor. Arch Acad Emerg Med. 2021;9(1):e9. pmid:33490966
  87. 87. Doron S, Ingalls R, Beauchamp A, Boehm J, Boucher H, Chow L, et al. Weekly SARS-CoV-2 screening of asymptomatic students and staff to guide and evaluate strategies for safer in-person learning. medRxiv. 2021 Mar 22.
  88. 88. Grossi E, Agnoli B, Baldini M, Illari S, Bonini R, Scagnelli G. Universal Sars-Cov-2 screening in pregnant women: experience from the Italian epidemic outbreak. Acta Biomed. 2021;92(S2):e2021001. pmid:33855978
  89. 89. Hicks SM, Pohl K, Neeman T, McNamara HA, Parsons KM, He JS, et al. A dual-antigen enzyme-linked immunosorbent assay allows the assessment of severe acute respiratory syndrome coronavirus 2 antibody seroprevalence in a low-transmission setting. J Infect Dis. 2021;223(1):10–4. pmid:33009908
  90. 90. Kiyasu Y, Akashi Y, Sugiyama A, Takeuchi Y, Notake S, Naito A, et al. A prospective evaluation of the analytical performance of GENECUBE HQ SARS-CoV-2 and GENECUBE FLU A/B. medRxiv. 2021 Feb 25. pmid:34003471
  91. 91. Lagi F, Trevisan S, Piccica M, Graziani L, Basile G, Mencarini J, et al. Use of the FebriDx point-of-care test for the exclusion of SARS-CoV-2 diagnosis in a population with acute respiratory infection during the second (COVID-19) wave in Italy. Int J Infect Dis. 2021;108:231–6. pmid:33901656
  92. 92. Latiano A, Tavano F, Panza A, Palmieri O, Niro GA, Andriulli N, et al. False positive results of IgM/IgG antibodies against antigen of the SARS-CoV-2 in sera stored before the 2020 Endemia in Italy. Int J Infect Dis. 2020;104:159–63. pmid:33383223
  93. 93. Li J, Hu X, Wang X, Yang J, Zhang L, Deng Q, et al. A novel one-pot rapid diagnostic technology for COVID-19. Anal Chim Acta. 2021;1154:338310. pmid:33736798
  94. 94. Marsic T, Ali Z, Tehseen M, Mahas A, Hamdan S, Mahfouz M. Vigilant: an engineered VirD2-Cas9 complex for lateral flow assay-based detection of SARS-CoV2. Nano Lett. 2021;21(8):3596–603. pmid:33844549
  95. 95. Schleicher T, Spenlinhauer T, Amadei M, Dasch N, Gordon J, Macleod G, et al. Development of a multiplexed synthetic control for rapid detection of SARS-CoV-2 and other respiratory pathogens using a nucleic acid syndromic testing panel. J Mol Diagnostics. 2020;22(11):S37.
  96. 96. Bello-Chavolla OY, Antonio-Villa NE, Fernández-Chirino L, Guerra E, Fermín-Martínez C, Márquez-Salinas A, et al. Diagnostic performance and clinical implications of rapid SARS-CoV-2 antigen testing in Mexico using real-world nationwide COVID-19 registry data. medRxiv. 2021 Jan 4.
  97. 97. Dalal A, Sonika U, Kumar M, George R, Kumar A, Srivastava S, et al. COVID-19 rapid antigen test: role in screening prior to gastrointestinal endoscopy. Clin Endosc. 2021 Mar 4. pmid:33657784
  98. 98. Downs LO, Eyre DW, O’Donnell D, Jeffery K. Home-based SARS-CoV-2 lateral flow antigen testing in hospital workers. J Infect. 2021;82(2):282–327. pmid:33573777
  99. 99. Haage VC, Moreira-Soto A, Sacks J, Corman V, Drosten C, Drexler JF. Limited specificity of SARS-CoV-2 antigen-detecting rapid diagnostic tests at low temperatures. medRxiv. 2021 Feb 3.
  100. 100. Hoehl S, Schenk B, Rudych O, Göttig S, Foppa I, Kohmer N, et al. At-home self-testing of teachers with a SARS-CoV-2 rapid antigen test to reduce potential transmissions in schools. medRxiv. 2020 Dec 7.
  101. 101. Marco A, Solé C, Abdo IJ, Turu E. Low sensitivity of rapid antigenic tests as a screening method in an outbreak of SARS-CoV-2 infection in prison. Enferm Infecc Microbiol Clin (Engl Ed). 2021 Feb 11. pmid:33707072
  102. 102. Matsuda EM, de Campos IB, de Oliveira IP, Colpas DR, Carmo A, Brígido LFM. Field evaluation of COVID-19 antigen tests versus RNA based detection: potential lower sensitivity compensated by immediate results, technical simplicity and low cost. J Med Virol. 2021;93(7):4405–10. pmid:33788270
  103. 103. Moreno G, Braun K, Pray I, Segaloff H, Lim A, Poulson K, et al. SARS-CoV-2 transmission in intercollegiate athletics not fully mitigated with daily antigen testing. medRxiv. 2021 Mar 6. pmid:33688665
  104. 104. Yamayoshi S, Sakai-Tagawa Y, Koga M, Akasaka O, Nakachi I, Koh H, et al. Comparison of rapid antigen tests for COVID-19. Viruses. 2020;12(12):1420. pmid:33322035
  105. 105. Yokota I, Shane P, Teshima T. Logistic advantage of two-step screening strategy for SARS-CoV-2 at airport quarantine. medRxiv. 2021 Jan 26. pmid:34174408
  106. 106. Häuser F, Sprinzl MF, Dreis KJ, Renzaho A, Youhanen S, Kremer WM, et al. Evaluation of a laboratory-based high-throughput SARS-CoV-2 antigen assay for non-COVID-19 patient screening at hospital admission. Med Microbiol Immunol. 2021;201(2–3):165–71. pmid:33856557
  107. 107. Colavita F, Vairo F, Meschi S, Valli MB, Lalle E, Castilletti C, et al. COVID-19 Rapid antigen test as screening strategy at points of entry: experience in Lazio region, central Italy, August–October 2020. Biomolecules. 2021;11(3):425. pmid:33805832
  108. 108. Kotsiou OS, Pantazopoulos I, Papagiannis D, Fradelos EC, Kanellopoulos N, Siachpazidou D, et al. Repeated antigen-based rapid diagnostic testing for estimating the coronavirus disease 2019 prevalence from the perspective of the workers’ vulnerability before and during the lockdown. Int J Environ Res Public Health. 2021;18(4):1638. pmid:33572118
  109. 109. Blairon L, Wilmet A, Beukinga I, Tre-Hardy M. Implementation of rapid SARS-CoV-2 antigenic testing in a laboratory without access to molecular methods: experiences of a general hospital. J Clin Virol. 2020;129:104472. pmid:32504944
  110. 110. Kashiwagi K, Ishii Y, Aoki K, Yagi S, Maeda T, Miyazaki T, et al. Immunochromatographic test for the detection of SARS-CoV-2 in saliva. medRxiv. 2020 May 25. pmid:33397587
  111. 111. Itoh K, Kawamitsu T, Osaka Y, Sato K, Suzuki Y, Kiriba C, et al. False positive results in severe acute respiratory coronavirus 2 (SARS-CoV-2) rapid antigen tests for inpatients. J Infect Chemother. 2021;27(7):1089–91. pmid:33810971
  112. 112. Prince-Guerra JL, Almendares O, Nolen LD, Gunn JKL, Dale AP, Buono SA, et al. Evaluation of Abbott BinaxNOW rapid antigen test for SARS-CoV-2 infection at two community-based testing sites—Pima County, Arizona, November 3–17, 2020. MMWR Morb Mortal Wkly Rep. 2021;70(3):100–5. pmid:33476316
  113. 113. Aoki K, Nagasawa T, Ishii Y, Yagi S, Kashiwagi K, Miyazaki T, et al. Evaluation of clinical utility of novel coronavirus antigen detection reagent, Espline SARS-CoV-2. J Infect Chemother. 2021;27(2):319–22. pmid:33388232
  114. 114. Caputo V, Bax C, Colantoni L, Peconi C, Termine A, Fabrizio C, et al. Comparative analysis of antigen and molecular tests for the detection of Sars-CoV-2 and related variants: a study on 4266 samples. Int J Infect Dis. 2021;108:187–9. pmid:33878460
  115. 115. Ford L, Lee C, Pray IW, Cole D, Bigouette JP, Abedi GR, et al. Epidemiologic characteristics associated with SARS-CoV-2 antigen-based test results, rRT-PCR cycle threshold values, subgenomic RNA, and viral culture results from university testing. Clin Infect Dis. 2021 Apr 13. pmid:33846714
  116. 116. Hirotsu Y, Maejima M, Shibusawa M, Nagakubo Y, Hosaka K, Amemiya K, et al. Comparison of automated SARS-CoV-2 antigen test for COVID-19 infection with quantitative RT-PCR using 313 nasopharyngeal swabs, including from seven serially followed patients. Int J Infect Dis. 2020;99:397–402. pmid:32800855
  117. 117. Ishii T, Sasaki M, Yamada K, Kato D, Osuka H, Aoki K, et al. Immunochromatography and chemiluminescent enzyme immunoassay for COVID-19 diagnosis. J Infect Chemother. 2021;27(6):915–8. pmid:33676843
  118. 118. Kiyasu Y, Takeuchi Y, Akashi Y, Kato D, Kuwahara M, Muramatsu S, et al. Prospective analytical performance evaluation of the QuickNavi-COVID19 Ag for asymptomatic individuals. medRxiv. 2021 Apr 7. pmid:34301485
  119. 119. Korenkov M, Poopalasingam N, Madler M, Vanshylla K, Eggeling R, Wirtz M, et al. Reliable assessment of in vitro SARS-CoV-2 infectivity by a rapid antigen detection test. medRxiv. 2021 Apr 6.
  120. 120. Kronberg Jakobsen K, Schmidt Jensen J, Todsen T, Lippert F, Martel CJ, Klokker M, et al. Detection of SARS-CoV-2 infection by rapid antigen test in comparison with RT-PCR in a public setting. medRxiv. 2021 Jan 25.
  121. 121. Kweon OJ, Lim YK, Kim HR, Choi Y, Kim MC, Choi SH, et al. Evaluation of rapid SARS-CoV-2 antigen tests, AFIAS COVID-19 Ag and ichroma COVID-19 Ag, with serial nasopharyngeal specimens from COVID-19 patients. PLoS ONE. 2021;16(4):e0249972. pmid:33831118
  122. 122. Landaas ET, Storm ML, Tollånes MC, Barlinn R, Kran AB, Bragstad K, et al. Diagnostic performance of a SARS-CoV-2 rapid antigen test in a large, Norwegian cohort. J Clin Virol. 2021;137:104789. pmid:33736946
  123. 123. McAulay K, Kaleta EJ, Grys TE. Rapid detection of SARS-CoV-2 antigen from serum in a hospitalized population. medRxiv. 2020 Dec 22.
  124. 124. McKay SL, Tobolowsky FA, Moritz ED, Hatfield KM, Bhatnagar A, LaVoie SP, et al. Performance evaluation of serial SARS-CoV-2 rapid antigen testing during a nursing home outbreak. Ann Intern Med. 2021;174(7):945–51. pmid:33900791
  125. 125. Pray IW, Ford L, Cole D, Lee C, Bigouette JP, Abedi GR, et al. Performance of an antigen-based test for asymptomatic and symptomatic SARS-CoV-2 testing at two university campuses—Wisconsin, September–October 2020. MMWR Morb Mortal Wkly Rep. 2021;69(5152):1642–7. pmid:33382679
  126. 126. Rastawicki W, Gierczyński R, Juszczyk G, Mitura K, Henry BM. Evaluation of PCL rapid point of care antigen test for detection of SARS-CoV-2 in nasopharyngeal swabs. J Med Virol. 2021;93(4):1920–2. pmid:33406271
  127. 127. Shah M, Salvatore P, Ford L, Kamitani E, Whaley M, Mitchell K, et al. Performance of repeat BinaxNOW SARS-CoV-2 antigen testing in a community setting, Wisconsin, November–December 2020. Clin Infect Dis. 2021;73(Suppl 1):S54–7. pmid:33909068
  128. 128. Uwamino Y, Nagata M, Aoki W, Nakagawa T, Inose R, Yokota H, et al. Accuracy of rapid antigen detection test for nasopharyngeal swab specimens and saliva samples in comparison with RT-PCR and viral culture for SARS-CoV-2 detection. J Infect Chemother. 2021;27(7):1058–62. pmid:33934920
  129. 129. Winkel BMF, Schram E, Gremmels H, Debast S, Schuurman R, Wensing AMJ, et al. Screening for SARS-CoV-2 infection in asymptomatic individuals using the Panbio COVID-19 Antigen Rapid Test (Abbott) compared to RT-qPCR. medRxiv. 2020 Dec 4.
  130. 130. Yamamoto K, Suzuki M, Yamada G, Sudo T, Nomoto H, Kinoshita N, et al. Utility of the antigen test for coronavirus disease 2019: factors influencing the prediction of the possibility of disease transmission. Int J Infect Dis. 2021;104:65–72. pmid:33401037
  131. 131. Ahava M, Kurkela S, Kuivanen S, Lappalainen M, Jarva H, Jaaskelainen A. Detection of SARS-CoV-2 nucleocapsid antigen from serum can aid in timing of COVID-19 infection. medRxiv. 2021 Jan 13.
  132. 132. Azzi L, Baj A, Alberio T, Lualdi M, Veronesi G, Carcano G, et al. Rapid salivary test suitable for a mass screening program to detect SARS-CoV-2: a diagnostic accuracy study. J Infect. 2020;81(3):E75–8. pmid:32579988
  133. 133. Barauna VG, Singh MN, Barbosa LL, Marcarini WD, Vassallo PF, Mill JG, et al. Ultrarapid on-site detection of SARS-CoV-2 infection using simple ATR-FTIR spectroscopy and an analysis algorithm: high sensitivity and specificity. Anal Chem. 2021;93(5):2950–8. pmid:33481583
  134. 134. Barlev-Gross M, Weiss S, Ben-Shmuel A, Sittner A, Eden K, Mazuz N, et al. Spike vs nucleocapsid SARS-CoV-2 antigen detection: application in nasopharyngeal swab specimens. Anal Bioanal Chem. 2021;413:3501–10. pmid:33768365
  135. 135. Cardozo KHM, Lebkuchen A, Okai GG, Schuch RA, Viana LG, Olive AN, et al. Establishing a mass spectrometry-based system for rapid detection of SARS-CoV-2 in large clinical sample cohorts. Nat Commun. 2020;11(1):6201. pmid:33273458
  136. 136. Cazares LH, Chaerkady R, Samuel Weng SH, Boo CC, Cimbro R, Hsu HE, et al. Development of a parallel reaction monitoring mass spectrometry assay for the detection of SARS-CoV-2 spike glycoprotein and nucleoprotein. Anal Chem. 2020;92(20):13813–21. pmid:32966064
  137. 137. Chen H, Li Z, Feng S, Wang A, Richard-Greenblatt M, Hutson E, et al. Femtomolar SARS-CoV-2 antigen detection using the microbubbling digital assay with smartphone readout enables antigen burden quantitation and dynamics tracking. medRxiv. 2021 Mar 26. pmid:33791710
  138. 138. Colavita F, Vairo F, Meschi S, Valli B, Lalle E, Castilletti C, et al. COVID-19 antigen rapid test as screening strategy at the points-of-entry: experience in Lazio region, central Italy, August–October 2020. medRxiv. 2020 Nov 30.
  139. 139. Conzelmann C, Gilg A, Groß R, Schütz D, Preising N, Ständker L, et al. An enzyme-based immunodetection assay to quantify SARS-CoV-2 infection. Antiviral Res. 2020;181:104882. pmid:32738255
  140. 140. Di Domenico M, De Rosa A, Boccellino M. Detection of SARS-COV-2 proteins using an ELISA test. Diagnostics (Basel). 2021;11(4):698. pmid:33919728
  141. 141. Diao B, Wen K, Zhang J, Chen J, Han C, Chen Y, et al. Accuracy of a nucleocapsid protein antigen rapid test in the diagnosis of SARS-CoV-2 infection. Clin Microbiol Infect. 2020;27(2):289.e1–4. pmid:33031947
  142. 142. Ducrest PJ. Development and evaluation of a new Swiss made SARS-CoV-2 antigen-detecting rapid test. medRxiv. 2021 Mar 26.
  143. 143. Grant BD, Anderson CE, Williford JR, Alonzo LF, Glukhova VA, Boyle DS, et al. SARS-CoV-2 coronavirus nucleocapsid antigen-detecting half-strip lateral flow assay toward the development of point of care tests using commercially available reagents. Anal Chem. 2020;92(16):11305–9. pmid:32605363
  144. 144. Huang L, Ding L, Zhou J, Chen S, Chen F, Zhao C, et al. One-step rapid quantification of SARS-CoV-2 virus particles via low-cost nanoplasmonic sensors in generic microplate reader and point-of-care device. Biosens Bioelectron. 2021;171:112685. pmid:33113383
  145. 145. Kyosei Y, Namba M, Yamura S, Takeuchi R, Aoki N, Nakaishi K, et al. Proposal of de novo antigen test for COVID-19: ultrasensitive detection of spike proteins of SARS-CoV-2. Diagnostics (Basel). 2020;10(8):594. pmid:32823866
  146. 146. Lee JH, Choi M, Jung Y, Lee SK, Lee CS, Kim J, et al. A novel rapid detection for SARS-CoV-2 spike 1 antigens using human angiotensin converting enzyme 2 (ACE2). Biosens Bioelectron. 2021;171:112715. pmid:33099241
  147. 147. Lee L, Liu F, Chen Y, Roma G. Quantitative and ultrasensitive in-situ immunoassay technology for SARS-CoV-2 detection in saliva. Research Square. 2021 Jan 18. pmid:33469572
  148. 148. Li Y, Peng Z, Holl NJ, Hassan MR, Pappas JM, Wei C, et al. MXene-graphene field-effect transistor sensing of influenza virus and SARS-CoV-2. ACS Omega. 2021;6(10):6643–53. pmid:33748577
  149. 149. Liu D, Ju C, Han C, Shi R, Chen X, Duan D, et al. Ultra-sensitive nanozyme-based chemiluminescence paper test for rapid diagnosis of SARS-CoV-2 infection. bioRxiv. 2020 Jun 5.
  150. 150. Mahari S, Roberts A, Shahdeo D, Gandhi S. eCovSens-ultrasensitive novel in-house built printed circuit board based electrochemical device for rapid detection of nCovid-19. bioRxiv. 2020 May 11.
  151. 151. Nash B, Badea A, Reddy A, Bosch M, Salcedo N, Gomez AR, et al. The impact of high frequency rapid viral antigen screening on COVID-19 spread and outcomes: a validation and modeling study. medRxiv. 2020 Nov 4.
  152. 152. Renuse S, Vanderboom P, Maus A, Kemp J, Gurtner K, Madugundu A, et al. Development of mass spectrometry-based targeted assay for direct detection of novel SARS-CoV-2 coronavirus from clinical specimens. medRxiv. 2020 Aug 6.
  153. 153. Ricks S, Kendall E, Dowdy D, Sacks J, Schumacher S, Arinaminpathy N. Quantifying the potential value of antigen-detection rapid diagnostic tests for COVID-19: a modelling analysis. BMC Med. 2020;19:75.
  154. 154. Seo G, Lee G, Kim MJ, Baek SH, Choi M, Ku KB, et al. Rapid detection of COVID-19 causative virus (SARS-CoV-2) in human nasopharyngeal swab specimens using field-effect transistor-based biosensor. ACS Nano. 2020;14(4):5135–42. pmid:32293168
  155. 155. Shao W, Shurin MR, Wheeler SE, He X, Star A. Rapid detection of SARS-CoV-2 antigens using high-purity semiconducting single-walled carbon nanotube-based field-effect transistors. ACS Appl Mater Interfaces. 2021;13(8):10321–7. pmid:33596036
  156. 156. Singh N, Ray P, Carlin A, Magallanes C, Morgan S, Laurent L, et al. Hitting the diagnostic sweet spot: point-of-care SARS-CoV-2 salivary antigen testing with an off-the-shelf glucometer. medRxiv. 2020 Oct 2.
  157. 157. Singh P, Chakraborty R, Marwal R, Radhakrishan VS, Bhaskar AK, Vashisht H, et al. A rapid and sensitive method to detect SARS-CoV-2 virus using targeted-mass spectrometry. J Proteins Proteom, 2020 Aug 31. pmid:33132628
  158. 158. Torrente-Rodríguez RM, Lukas H, Tu J, Min J, Yang Y, Xu C, et al. SARS-CoV-2 RapidPlex: a graphene-based multiplexed telemedicine platform for rapid and low-cost COVID-19 diagnosis and monitoring. Matter. 2020;3(6):1981–98. pmid:33043291
  159. 159. Vadlamani BS, Uppal T, Verma S, Misra M. Functionalized TiO2 nanotube-based electrochemical biosensor for rapid detection of SARS-CoV-2. Sensors. 2020;20(20):5871. pmid:33080785
  160. 160. Wang H, Hogan CA, Verghese M, Solis D, Sibai M, Huang C, et al. Ultra-sensitive severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antigen detection for the diagnosis of coronavirus disease 2019 (COVID-19) in upper respiratory samples. Clin Infect Dis. 2021 Apr 8. pmid:33830203
  161. 161. Yakoh A, Pimpitak U, Rengpipat S, Hirankarn N, Chailapakul O, Chaiyo S. Paper-based electrochemical biosensor for diagnosing COVID-19: detection of SARS-CoV-2 antibodies and antigen. Biosens Bioelectron. 2021;176:112912. pmid:33358057
  162. 162. Zakashansky J, Imamura A, Salgado D, Romero Mercieca H, Aguas RFL, Lao A, et al. Detection of the SARS-CoV-2 spike protein in saliva with Shrinky-Dink electrodes. medRxiv. 2020 Nov 18. pmid:33236028
  163. 163. Zhang CY, Zhou L, Du K, Zhang Y, Wang J, Chen LJ, et al. Foundation and clinical evaluation of a new method for detecting SARS-CoV-2 antigen by fluorescent microsphere immunochromatography. Front Cell Infect Microbiol. 2020;10:553837. pmid:33330119
  164. 164. Zou M, Su F, Zhang R, Jiang X, Xiao H, Yan X, et al. rapid point-of-care testing for SARS-CoV-2 virus nucleic acid detection by an isothermal and nonenzymatic signal amplification system coupled with a lateral flow immunoassay strip. Sens Actuators B Chem. 2021;342:129899. pmid:33840901
  165. 165. Akingba OL, Sprong K, Hardie DR. Field performance evaluation of the PanBio rapid SARS-CoV-2 antigen assay in an epidemic driven by 501Y.v2 (lineage B.1.351) in the Eastern Cape, South Africa. J Clin Virol Plus. 2021;1(1–2):100013.
  166. 166. Asai N, Sakanashi D, Ohashi W, Nakamura A, Kawamoto Y, Miyazaki N, et al. Efficacy and validity of automated quantitative chemiluminescent enzyme immunoassay for SARS-CoV-2 antigen test from saliva specimen in the diagnosis of COVID-19. J Infect Chemother. 2021;27(7):1039–42. pmid:33840598
  167. 167. Baro B, Rodo P, Ouchi D, Bordoy A, Saya Amaro E, Salsench S, et al. Performance characteristics of five antigen-detecting rapid diagnostic test (Ag-RDT) for SARS-CoV-2 asymptomatic infection: a head-to-head benchmark comparison. J Infect. 2021;82(6):269–75. pmid:33882299
  168. 168. Basso D, Aita A, Padoan A, Cosma C, Navaglia F, Moz S, et al. Salivary SARS-CoV-2 antigen rapid detection: a prospective cohort study. Clin Chim Acta. 2020;517:54–9. pmid:33626369
  169. 169. Bruzzone B, De Pace V, Caligiuri P, Ricucci V, Guarona G, Pennati BM, et al. Comparative diagnostic performance of different rapid antigen detection tests for COVID-19 in the real-world hospital setting. Int J Infect Dis. 2021;107:215–8. pmid:33930540
  170. 170. Caruana G, Croxatto A, Kampouri E, Kritikos A, Opota O, Foerster M, et al. ImplemeNting SARS-CoV-2 Rapid antigen testing in the Emergency wArd of a Swiss univErsity hospital: the INCREASE study. Microorganisms. 2021;9(4):798. pmid:33920307
  171. 171. Caruana G, Lebrun L-L, Aebischer O, Opota O, Urbano L, DeRham M, et al. The dark side of SARS-CoV-2 rapid antigen testing: screening asymptomatic patients. medRxiv. 2021 Apr 27. pmid:34007453
  172. 172. Ciotti M, Maurici M, Pieri M, Andreoni M, Bernardini S. Performance of a rapid antigen test in the diagnosis of SARS-CoV-2 infection. J Med Virol. 2021;93:2988–91. pmid:33527409
  173. 173. Cubas Atienzar A, Kontogianni K, Edwards T, Wooding D, Buist K, Thompson C, et al. Limit of detection in different matrices of nineteen commercially available rapid antigen tests for the detection of SARS-CoV-2. medRxiv. 2021 Mar 22.
  174. 174. Del Vecchio C, Brancaccio G, Brazzale AR, Lavezzo E, Onelia F, Franchin E, et al. Emergence of N antigen SARS-CoV-2 genetic variants escaping detection of antigenic tests. medRxiv. 2021 Mar 26.
  175. 175. Domínguez Fernández M, Peña Rodríguez MF, Lamelo Alfonsín F, Bou Arévalo G. Experience with Panbio rapid antigens test device for the detection of SARS-CoV-2 in nursing homes. Enferm Infecc Microbiol Clin (Engl Ed). 2021 Jan 19. pmid:33551148
  176. 176. Drain PK, Ampajwala M, Chappel C, Gvozden AB, Hoppers M, Wang M, et al. A rapid, high-sensitivity SARS-CoV-2 nucleocapsid immunoassay to aid diagnosis of acute COVID-19 at the point of care: a clinical performance study. Infect Dis Ther. 2021;10(2):753–61. pmid:33629225
  177. 177. Faíco-Filho KS, Finamor Júnior FE, Moreira LVL, Lins PRG, Justo AFO, Bellei N. Evaluation of the Panbio COVID-19 Ag Rapid Test at an emergency room in a hospital in São Paulo, Brazil. medRxiv. 2021 Mar 24.
  178. 178. Favresse J, Gillot C, Oliveira M, Cadrobbi J, Elsen M, Eucher C, et al. Head-to-head comparison of rapid and automated antigen detection tests for the diagnosis of SARS-CoV-2 infection. J Clin Med. 2021;10(2):265. pmid:33450853
  179. 179. Ferguson J, Dunn S, Best A, Mirza J, Percival B, Mayhew M, et al. Validation testing to determine the sensitivity of lateral flow testing for asymptomatic SARS-CoV-2 detection in low prevalence settings: testing frequency and public health messaging is key. PLoS Biol. 2021;19(4):e3001216. pmid:33914730
  180. 180. Filgueiras P, Corsini C, Almeida NBF, Assis J, Pedrosa ML, de Oliveira A, et al. COVID-19 rapid antigen test at hospital admission associated to the knowledge of individual risk factors allow overcoming the difficulty of managing suspected patients in hospitals COVID-19 rapid antigen test facilitates the management of suspected patients on hospital admission. medRxiv. 2021 Jan 8.
  181. 181. Foundation for Innovative New Diagnostics. FIND evaluation of Boditech Medical, Inc. iChroma COVID-19 Ag Test. External Report Version 10, 23 February 2021. Geneva: Foundation for Innovative New Diagnostics; 2021.
  182. 182. Foundation for Innovative New Diagnostics. FIND evaluation of Joysbio (Tianjin) Biotechnology Co., Ltd. SARS-CoV-2 Antigen Rapid Test Kit (Colloidal Gold). External Report Version 10, 11 February 2021. Geneva: Foundation for Innovative New Diagnostics; 2021.
  183. 183. Foundation for Innovative New Diagnostics. FIND evaluation of Guangzhou Wondfo Biotech Co., Ltd Wondfo 2019-nCoV Antigen Test (Lateral Flow Method). Public Report Version 10, 25 February 2021. Geneva: Foundation for Innovative New Diagnostics; 2021.
  184. 184. Foundation for Innovative New Diagnostics. FIND evaluation of Abbott Panbio COVID-19 Ag Rapid Test Device (NASAL). External Report Version 10. Geneva: Foundation for Innovative New Diagnostics; 11 February 2021, 2021.
  185. 185. Foundation for Innovative New Diagnostics. FIND evaluation of Bionote, Inc. NowCheck COVID-19 Ag Test, nasal swab. External Report Version 10, 30 March 2021. Geneva: Foundation for Innovative New Diagnostics; 2021.
  186. 186. Foundation for Innovative New Diagnostics. FIND evaluation of Fujirebio Inc. Espline SARS-CoV-2. External Report Version 10, 29 March 2021. Geneva: Foundation for Innovative New Diagnostics; 2021.
  187. 187. Foundation for Innovative New Diagnostics. FIND evaluation of Mologic Ltd, COVID 19 RAPID ANTIGEN TEST. External Report Version 10, 23 April 2021. Geneva: Foundation for Innovative New Diagnostics; 2021.
  188. 188. Foundation for Innovative New Diagnostics. FIND evaluation of NADAL COVID-19 Ag Rapid Test. External Report Version 10, 26 April 2021. Geneva: Foundation for Innovative New Diagnostics; 2021.
  189. 189. Gili A, Paggi R, Russo C, Cenci E, Pietrella D, Graziani A, et al. Evaluation of automated test Lumipulse G SARS-CoV-2 antigen assay for detection of SARS-CoV-2 nucleocapsid protein (NP) in nasopharyngeal swabs for community and population screening. Int J Infect Dis. 2021;105:P391–6. pmid:33647511
  190. 190. Gomez Marti JLG, Gribschaw J, McCullough M, Mallon A, Acero J, Kinzler A, et al. Differences in detected viral loads guide use of SARS-CoV-2 antigen-detection assays towards symptomatic college students and children. medRxiv. 2021 Feb 1.
  191. 191. Haage V, Ferreira de Oliveira-Filho E, Moreira-Soto A, Kühne A, Fischer C, Sacks JA, et al. Impaired performance of SARS-CoV-2 antigen-detecting rapid diagnostic tests at elevated and low temperatures. J Clin Virol. 2021;138:104796. pmid:33773413
  192. 192. Halfon P, Penaranda G, Khiri H, Garcia V, Drouet H, Philibert P, et al. An optimized stepwise algorithm combining rapid antigen and RT-qPCR for screening of COVID-19 patients. medRxiv. 2021 Jan 15.
  193. 193. Hirotsu Y, Maejima M, Shibusawa M, Amemiya K, Nagakubo Y, Hosaka K, et al. Prospective study of 1,308 nasopharyngeal swabs from 1,033 patients using the LUMIPULSE SARS-CoV-2 antigen test: comparison with RT-qPCR. Int J Infect Dis. 2021;105:7–14. pmid:33556612
  194. 194. Homza M, Zelena H, Janosek J, Tomaskova H, Jezo E, Kloudova A, et al. Five antigen tests for SARS-CoV-2: virus viability matters. Viruses. 2021;13(4):684. pmid:33921164
  195. 195. Houston H, Gupta-Wright A, Toke-Bjolgerud E, Biggin-Lamming J, John L. Diagnostic accuracy and utility of SARS-CoV-2 antigen lateral flow assays in medical admissions with possible COVID-19. J Hosp Infect. 2021;110:203–5. pmid:33539935
  196. 196. Jääskeläinen AE, Ahava MJ, Jokela P, Szirovicza L, Pohjala S, Vapalahti O, et al. Evaluation of three rapid lateral flow antigen detection tests for the diagnosis of SARS-CoV-2 infection. J Clin Virol. 2021;137:104785. pmid:33711694
  197. 197. James AE, Gulley T, Kothari A, Holder K, Garner K, Patil N. Performance of the BinaxNOW COVID-19 Antigen Card test relative to the SARS-CoV-2 real-time reverse transcriptase polymerase chain reaction assay among symptomatic and asymptomatic healthcare employees. Infect Control Hosp Epidemiol. 2021 Jan 25. pmid:33487197
  198. 198. Jungnick S, Hobmaier B, Mautner L, Hoyos M, Haase M, Baiker A, et al. Detection of the new SARS-CoV-2 variants of concern B.1.1.7 and B.1.351 in five SARS-CoV-2 rapid antigen tests (RATs), Germany, March 2021. Euro Surveill. 2021;26(16):2100413. pmid:33890568
  199. 199. Kannian P, Lavanya C, Ravichandran K, Gita JB, Mahanathi P, Ashwini V, et al. SARS-CoV2 antigen in whole mouth fluid may be a reliable rapid detection tool. Oral Dis. 2021 Feb 3. pmid:33534926
  200. 200. Kenyeres B, Ánosi N, Bányai K, Mátyus M, Orosz L, Kiss A, et al. Comparison of four PCR and two point of care assays used in the laboratory detection of SARS-CoV-2. J Virol Methods. 2021;293:114165. pmid:33872650
  201. 201. Kernéis S, Elie C, Fourgeaud J, Choupeaux L, Delarue SM, Alby M-L, et al. Accuracy of antigen and nucleic acid amplification testing on saliva and naopharyngeal samples for detection of SARS-CoV-2 in ambulatory care. medRxiv. 2021 Apr 11.
  202. 202. Kilic A, Hiestand B, Palavecino E. Evaluation of performance of the BD Veritor SARS-CoV-2 chromatographic immunoassay test in patients with symptoms of COVID-19. J Clin Microbiol. 2021;59(5):e00260–21. pmid:33637583
  203. 203. Kohmer N, Toptan T, Pallas C, Karaca O, Pfeiffer A, Westhaus S, et al. The comparative clinical performance of four SARS-CoV-2 rapid antigen tests and their correlation to infectivity in vitro. J Clin Med. 2021;10(2):328. pmid:33477365
  204. 204. Krüger L, Klein JAF, Tobian F, Gaeddert M, Lainati F, Klemm S, et al. Evaluation of accuracy, exclusivity, limit-of-detection and ease-of-use of LumiraDx-Antigen-detecting point-of-care device for SARS-CoV-2. medRxiv. 2021 Mar 5.
  205. 205. L’Huillier A, Lacour M, Sadiku D, Gadiri M, De Siebenthal L, Schibler M, et al. Diagnostic accuracy of SARS-CoV-2 rapid antigen detection testing in symptomatic and asymptomatic children in the clinical setting. medRxiv. 2021 Apr 20. pmid:34190574
  206. 206. Lefever S, Indevuyst C, Cuypers L, Dewaele K, Yin N, Cotton F, et al. Comparison of the quantitative DiaSorin Liaison antigen test to RT-PCR for the diagnosis of COVID-19 in symptomatic and asymptomatic outpatients. J Clin Microbiol. 2021;59(7):e0037421. pmid:33849953
  207. 207. Lindner A, Nikolai O, Rohardt C, Kausch F, Wintel M, Gertler M, et al. SARS-CoV-2 patient self-testing with an antigen-detecting rapid test: a head-to-head comparison with professional testing. medRxiv. 2021 Jan 8.
  208. 208. Mak GCK, Lau SSY, Wong KKY, Chow NLS, Lau CS, Lam ETK, et al. Evaluation of rapid antigen detection kit from the WHO Emergency Use List for detecting SARS-CoV-2. J Clin Virol. 2021;134:104712. pmid:33338894
  209. 209. Mboumba Bouassa RS, Veyer D, Péré H, Bélec L. Analytical performances of the point-of-care SIENNA COVID-19 Antigen Rapid Test for the detection of SARS-CoV-2 nucleocapsid protein in nasopharyngeal swabs: a prospective evaluation during the COVID-19 second wave in France. Int J Infect Dis. 2021;106:8–12. pmid:33746093
  210. 210. Menchinelli G, Bordi L, Marzialiotti F, Palucci I, Capobianchi M, Sberna G, et al. Lumipulse G SARS-CoV-2 Ag Assay evaluation for SARS-CoV-2 antigen detection using 594 nasopharyngeal swab samples from different testing groups. Clin Chem Lab Med. 2021;59(8):1468–76. pmid:33823089
  211. 211. Micocci M, Buckle P, Hayward G, Allen J, Davies K, Kierkegaard P, et al. Point of care testing using rapid automated antigen testing for SARS-COV-2 in care homes—an exploratory safety, usability and diagnostic agreement evaluation. medRxiv. 2021 Apr 26.
  212. 212. Möckel M, Corman VM, Stegemann MS, Hofmann J, Stein A, Jones TC, et al. SARS-CoV-2 antigen rapid immunoassay for diagnosis of COVID-19 in the emergency department. Biomarkers. 2021;26(3):213–20. pmid:33455451
  213. 213. Muhi S, Tayler N, Hoang T, Ballard SA, Graham M, Rojek A, et al. Multi-site assessment of rapid, point-of-care antigen testing for the diagnosis of SARS-CoV-2 infection in a low-prevalence setting: a validation and implementation study. Lancet Reg Health West Pac. 2021;9:100115. pmid:33937887
  214. 214. Ngo Nsoga M-T, Kronig I, Perez Rodriguez FJ, Sattonnet-Roche P, Da Silva D, Helbling J, et al. Diagnostic accuracy of Panbio rapid antigen tests on oropharyngeal swabs for detection of SARS-CoV-2. medRxiv. 2021 Feb 1. pmid:34166410
  215. 215. Nikolai O, Rohardt C, Tobian F, Junge A, Corman V, Jones T, et al. Anterior nasal versus nasal mid-turbinate sampling for a SARS-CoV-2 antigen-detecting rapid test: does localisation or professional collection matter? medRxiv. 2021 Feb 16.
  216. 216. Nörz D, Olearo F, Perisic S, Bauer M, Riester E, Schneider T, et al. Multicenter evaluation of a fully automated high-throughput SARS-CoV-2 antigen immunoassay. medRxiv. 2021 Apr 15.
  217. 217. Okoye NC, Barker AP, Curtis K, Orlandi RR, Snavely EA, Wright C, et al. Performance characteristics of BinaxNOW COVID-19 Antigen Card for screening asymptomatic individuals in a university setting. J Clin Microbiol. 2021;59(4):e03282–20. pmid:33509809
  218. 218. Osmanodja B, Budde K, Zickler D, Naik M, Hofmann J, Gertler M, et al. Diagnostic accuracy of a novel SARS-CoV-2 antigen-detecting rapid diagnostic test from standardized self-collected anterior nasal swabs. medRxiv. 2021 Apr 23. pmid:34068236
  219. 219. Osterman A, Baldauf HM, Eletreby M, Wettengel JM, Afridi SQ, Fuchs T, et al. Evaluation of two rapid antigen tests to detect SARS-CoV-2 in a hospital setting. Med Microbiol Immunol. 2021;210(1):65–72. pmid:33452927
  220. 220. Pena M, Ampuero M, Garces C, Gaggero A, Garcia P, Velasquez MS, et al. Performance of SARS-CoV-2 rapid antigen test compared with real-time RT-PCR in asymptomatic individuals. medRxiv. 2021 Feb 13. pmid:33945868
  221. 221. Pérez-García F, Romanyk J, Gómez-Herruz P, Arroyo T, Pérez-Tanoira R, Linares M, et al. Diagnostic performance of CerTest and Panbio antigen rapid diagnostic tests to diagnose SARS-CoV-2 infection. J Clin Virol. 2021;137:104781. pmid:33639492
  222. 222. Peto T, UK COVID-19 Lateral Flow Oversight Team. COVID-19: rapid antigen detection for SARS-CoV-2 by lateral flow assay: a national systematic evaluation for mass-testing. medRxiv. 2021 Jan 26. pmid:34101770
  223. 223. Pickering S, Batra R, Snell L, Merrick B, Nebbia G, Douthwaite S, et al. Comparative performance of SARS-CoV-2 lateral flow antigen tests demonstrates their utility for high sensitivity detection of infectious virus in clinical specimens. medRxiv. 2021 Mar 2.
  224. 224. Pollock N, Jacobs J, Tran K, Cranston A, Smith S, O’Kane C, et al. Performance and implementation evaluation of the Abbott BinaxNOW Rapid Antigen Test in a high-throughput drive-through community testing site in Massachusetts. J Clin Microbiol. 2021;59(5):e00083–21. pmid:33622768
  225. 225. Pollock N, Tran K, Jacobs J, Cranston A, Smith S, O’Kane C, et al. Performance and operational evaluation of the Access Bio CareStart Rapid Antigen Test in a high-throughput drive-through community testing site in Massachusetts. medRxiv. 2021 Mar 9.
  226. 226. Ristić M, Nikolić N, Čabarkapa V, Turkulov V, Petrović V. Validation of the STANDARD Q COVID-19 antigen test in Vojvodina, Serbia. PLoS ONE. 2021;16(2):e0247606. pmid:33617597
  227. 227. Rodgers M, Batra R, Snell L, Daghfal D, Roth R, Huang S, et al. Detection of SARS-CoV-2 variants by Abbott molecular, antigen, and serological tests. medRxiv. 2021 Apr 26.
  228. 228. Rottenstreich A, Zarbiv G, Kabiri D, Porat S, Sompolinsky Y, Reubinoff B, et al. Rapid antigen detection testing for universal screening for severe acute respiratory syndrome coronavirus 2 in women admitted for delivery. Am J Obstet Gynecol. 2021;224(5):539–40. pmid:33453181
  229. 229. Salvagno GL, Gianfilippi G, Bragantini D, Henry BM, Lippi G. Clinical assessment of the Roche SARS-CoV-2 rapid antigen test. Diagnosis (Berl). 2021 Jan 18. pmid:33554511
  230. 230. Sberna G, Lalle E, Capobianchi MR, Bordi L, Amendola A. Letter of concern re: “Immunochromatographic test for the detection of SARS-CoV-2 in saliva. J Infect Chemother. 2021 Feb;27(2):384–386. 10.1016/j.jiac.2020.11.016”. J Infect Chemother. 2021;27(7):1129–30. pmid:33888419
  231. 231. Schuit E, Veldhuijzen IK, Venekamp RP, van den Bijllaardt W, Pas SD, Lodder EB, et al. Diagnostic accuracy of rapid antigen tests in pre-/asymptomatic close contacts of individuals with a confirmed SARS-CoV-2 infection. medRxiv. 2021 Mar 23.
  232. 232. Seitz T, Schindler S, Winkelmeyer P, Zach B, Wenisch C, Zoufaly A, et al. Evaluation of rapid antigen tests based on saliva for the detection of SARS-CoV-2. J Med Virol. 2021;93(7):4161–2. pmid:33788280
  233. 233. Shidlovskaya E, Kuznetsova N, Divisenko E, Nikiforova M, Siniavin A, Ogarkova D, et al. The value of rapid antigen tests to identify carriers of viable SARS-CoV-2. medRxiv. 2021 Mar 12.
  234. 234. Stohr JJM, Zwart VF, Goderski G, Meijer A, Nagel-Imming RS, Kluytmans-van den Bergh MFQ, et al. Self-testing for the detection of SARS-CoV-2 infection with rapid antigen tests. medRxiv. 2021 Feb 23.
  235. 235. Stokes W, Berenger BM, Portnoy D, Scott B, Szelewicki J, Singh T, et al. Clinical performance of the Abbott Panbio with nasopharyngeal, throat, and saliva swabs among symptomatic individuals with COVID-19. Eur J Clin Microbiol Infect Dis. 2021;40(8):1721–6. pmid:33742322
  236. 236. Strömer A, Rose R, Schäfer M, Schön F, Vollersen A, Lorentz T, et al. Performance of a point-of-care test for the rapid detection of SARS-CoV-2 antigen. Microorganisms. 2020;9(1):58. pmid:33379279
  237. 237. Takeuchi Y, Akashi Y, Kato D, Kuwahara M, Muramatsu S, Ueda A, et al. The evaluation of a newly developed antigen test (QuickNavi-COVID19 Ag) for SARS-CoV-2: a prospective observational study in Japan. J Infect Chemother. 2021;27(6):890–4. pmid:33727026
  238. 238. Takeuchi Y, Akashi Y, Kato D, Kuwahara M, Muramatsu S, Ueda A, et al. Diagnostic performance and characteristics of anterior nasal collection for the SARS-CoV-2 antigen test: a prospective study. Sci Rep. 2021;11:10519. pmid:34006975
  239. 239. Thakur P, Saxena S, Manchanda V, Rana N, Goel R, Arora R. Utility of antigen-based rapid diagnostic test for detection of SARS-CoV-2 virus in routine hospital settings. Lab Med. 2021 Apr 30. pmid:33928384
  240. 240. Thell R, Kallab V, Weinhappel W, Mueckstein W, Heschl L, Heschl M, et al. Evaluation of a novel, rapid antigen detection test for the diagnosis of SARS-CoV-2. medRxiv. 2021 Apr 22.
  241. 241. Torres I, Poujois S, Albert E, Álvarez G, Colomina J, Navarro D. Point-of-care evaluation of a rapid antigen test (CLINITEST® Rapid COVID-19 Antigen Test) for diagnosis of SARS-CoV-2 infection in symptomatic and asymptomatic individuals. J Infect. 2021;82(5):e11–2. pmid:33587922
  242. 242. Turcato G, Zaboli A, Pfeifer N, Ciccariello L, Sibilio S, Tezza G, et al. Clinical application of a rapid antigen test for the detection of SARS-CoV-2 infection in symptomatic and asymptomatic patients evaluated in the emergency department: a preliminary report. J Infect. 2020;82:E14–6. pmid:33347944
  243. 243. Villaverde S, Domínguez-Rodríguez S, Sabrido G, Pérez-Jorge C, Plata M, Romero MP, et al. Diagnostic accuracy of the Panbio SARS-CoV-2 Antigen Rapid Test compared with RT-PCR testing of nasopharyngeal samples in the pediatric population. J Pediatr. 2021;232:P287–9.E4. pmid:33484697
  244. 244. Wagenhäuser I, Knies K, Rauschenberger V, Eisenmann M, McDonogh M, Petri N, et al. Clinical performance evaluation of SARS-CoV-2 rapid antigen testing in point of care usage in comparison to RT-qPCR. medRxiv. 2021 Mar 29. pmid:34186490
  245. 245. Yin N, Debuysschere C, Decroly M, Bouazza FZ, Collot V, Martin C, et al. SARS-CoV-2 diagnostic tests: algorithm and field evaluation from the near patient testing to the automated diagnostic platform. Front Med. 2021;8:380. pmid:33889587
  246. 246. Young BC, Eyre DW, Jeffery K. Use of lateral flow devices allows rapid triage of patients with SARS-CoV-2 on admission to hospital. J Infect. 2021;82:276–316. pmid:33662408
  247. 247. Abdulrahman A, Mustafa F, AlAwadhi AI, Alansari Q, AlAlawi B, AlQahtani M. Comparison of SARS-COV-2 nasal antigen test to nasopharyngeal RT-PCR in mildly symptomatic patients. medRxiv. 2020 Dec 8.
  248. 248. Agulló V, Fernández-González M, Ortiz de la Tabla V, Gonzalo-Jiménez N, García JA, Masiá M, et al. Evaluation of the rapid antigen test Panbio COVID-19 in saliva and nasal swabs: a population-based point-of-care study. J Infect. 2021;82(5):186–230. pmid:33309541
  249. 249. Albert E, Torres I, Bueno F, Huntley D, Molla E, Fernandez-Fuentes MA, et al. Field evaluation of a rapid antigen test (Panbio COVID-19 Ag Rapid Test Device) for COVID-19 diagnosis in primary healthcare centres. Clin Microbiol Infect. 2020;27(3):472.e7–10. pmid:33189872
  250. 250. Alemany A, Baro B, Ouchi D, Ubals M, Corbacho-Monné M, Vergara-Alert J, et al. Analytical and clinical performance of the Panbio COVID-19 antigen-detecting rapid diagnostic test. J Infect. 2020;82(5):186–230. pmid:33421447
  251. 251. Beck ET, Paar W, Fojut L, Serwe J, Jahnke RR. Comparison of Quidel Sofia SARS FIA Test to Hologic Aptima SARS-CoV-2 TMA Test for diagnosis of COVID-19 in symptomatic outpatients. J Clin Microbiol. 2020;59(2):e02727–20. pmid:33239376
  252. 252. Berger A, Ngo Nsoga M-T, Perez Rodriguez FJ, Abi Aad Y, Sattonnet P, Gayet-Ageron A, et al. Diagnostic accuracy of two commercial SARS-CoV-2 antigen-detecting rapid tests at the point of care in community-based testing centers. PLoS ONE. 2020;16(3):e0248921. pmid:33788882
  253. 253. Bulilete O, Lorente P, Leiva A, Carandell E, Oliver A, Rojo E, et al. Evaluation of the Panbio rapid antigen test for SARS-CoV-2 in primary health care centers and test sites. medRxiv. 2020 Nov 16.
  254. 254. Cerutti F, Burdino E, Milia MG, Allice T, Gregori G, Bruzzone B, et al. Urgent need of rapid tests for SARS CoV-2 antigen detection: evaluation of the SD-Biosensor antigen test for SARS-CoV-2. J Clin Virol. 2020;132:104654. pmid:33053494
  255. 255. Chaimayo C, Kaewnaphan B, Tanlieng N, Athipanyasilp N, Sirijatuphat R, Chayakulkeeree M, et al. Rapid SARS-CoV-2 antigen detection assay in comparison with real-time RT-PCR assay for laboratory diagnosis of COVID-19 in Thailand. Virol J. 2020;17(1):177. pmid:33187528
  256. 256. Corman V, Haage V, Bleicker T, Schmidt ML, Muehlemann B, Zuchowski M, et al. Comparison of seven commercial SARS-CoV-2 rapid point-of-care antigen tests. Lancet Microbe. 2021;2(7):e311–9. pmid:33846704
  257. 257. Drevinek P, Hurych J, Kepka Z, Briksi A, Kulich M, Zajac M, et al. The sensitivity of SARS-CoV-2 antigen tests in the view of large-scale testing. medRxiv. 2020 Nov 24.
  258. 258. Fenollar F, Bouam A, Ballouche M, Fuster L, Prudent E, Colson P, et al. Evaluation of the Panbio Covid-19 rapid antigen detection test device for the screening of patients with Covid-19. J Clin Microbiol. 2020;59:e02589–20. pmid:33139420
  259. 259. Foundation for Innovative New Diagnostics. FIND evaluation of Bionote, Inc. NowCheck COVID-19 Ag Test. External Report Version 15, 20 April 2021. Geneva: Foundation for Innovative New Diagnostics; 2020.
  260. 260. Foundation for Innovative New Diagnostics. FIND evaluation of RapiGEN Inc. BIOCREDIT COVID-19 Ag. External Report Version 21, 10 December 2020. Geneva: Foundation for Innovative New Diagnostics; 2020.
  261. 261. Foundation for Innovative New Diagnostics. FIND evaluation of SD Biosensor, Inc. STANDARD™ F COVID-19 Ag FIA. External Report Version 21, 10 December 2020. Geneva: Foundation for Innovative New Diagnostics; 2020.
  262. 262. Foundation for Innovative New Diagnostics. FIND evaluation of SD Biosensor, Inc. STANDARD Q COVID-19 Ag Test. External Report Version 21, 10 December 2020. Geneva: Foundation for Innovative New Diagnostics; 2020.
  263. 263. Gremmels H, Winkela BMF, Schuurmana R, Rosinghb A, Rigterc NAM, Rodriguezd O, et al. Real-life validation of the Panbio COVID-19 Antigen Rapid Test (Abbott) in community-dwelling subjects with symptoms of potential SARS-CoV-2 infection. EClinicalMedicine. 2020;31:100677. pmid:33521610
  264. 264. Gupta A, Khurana S, Das R, Srigyan D, Singh A, Mittal A, et al. Rapid chromatographic immunoassay-based evaluation of COVID-19: a cross-sectional, diagnostic test accuracy study & its implications for COVID-19 management in India. Indian J Med Res. 2020;153(1):126. pmid:33818469
  265. 265. Herrera V, Hsu V, Adewale A, Hendrix T, Johnson L, Kuhlman J, et al. Testing of healthcare workers exposed to COVID19 with rapid antigen detection. medRxiv. 2020 Aug 18.
  266. 266. Iglὁi Z, Velzing J, van Beek J, van de Vijver D, Aron G, Ensing R, et al. Clinical evaluation of the Roche/SD Biosensor rapid antigen test with symptomatic, non-hospitalized patients in a municipal health service drive-through testing site. Emerg Infect Dis. 2020;27(5):1323–9. pmid:33724916
  267. 267. Krüger LJ, Gaeddert M, Tobian F, Lainati F, Gottschalk C, Klein JAF, et al. Evaluation of the accuracy and ease-of-use of Abbott PanBio—a WHO emergency use listed, rapid, antigen-detecting point-of-care diagnostic test for SARS-CoV-2. medRxiv. 2020 Dec d.
  268. 268. Krüttgen A, Cornelissen CG, Dreher M, Hornef MW, Imöhl M, Kleinesa M. Comparison of the SARS-CoV-2 rapid antigen test to the real star Sars-CoV-2 RT PCR kit. J Virol Methods. 2020;288:114024. pmid:33227341
  269. 269. Lambert-Niclot S, Cuffel A, Le Pape S, Vauloup-Fellous C, Morand-Joubert L, Roque-Afonso AM, et al. Evaluation of a rapid diagnostic assay for detection of SARS-CoV-2 antigen in nasopharyngeal swabs. J Clin Microbiol. 2020;58(8):e00977–20. pmid:32404480
  270. 270. Linares M, Pérez-Tanoira R, Carrero A, Romanyk J, Pérez-García F, Gómez-Herruz P, et al. Panbio antigen rapid test is reliable to diagnose SARS-CoV-2 infection in the first 7 days after the onset of symptoms. J Clin Virol. 2020;133:104659. pmid:33160179
  271. 271. Lindner A, Nikolai O, Rohardt C, Burock S, Hülso C, Bölke A, et al. Head-to-head comparison of SARS-CoV-2 antigen-detecting rapid test with professional-collected nasal versus nasopharyngeal swab. Eur Respir J. 2020;57(5):2004430.
  272. 272. Lindner AK, Nikolai O, Kausch F, Wintel M, Hommes F, Gertler M, et al. Head-to-head comparison of SARS-CoV-2 antigen-detecting rapid test with self-collected nasal swab versus professional-collected nasopharyngeal swab. Eur Respir J. 2021;57(4):2003961. pmid:33303544
  273. 273. Liotti FM, Menchinelli G, Lalle E, Palucci I, Marchetti S, Colavita F, et al. Performance of a novel diagnostic assay for rapid SARS-CoV-2 antigen detection in nasopharynx samples. Clin Microbiol Infect. 2020;27:487–8. pmid:32979567
  274. 274. Mak GC, Lau SS, Wong KK, Chow NL, Lau CS, Lam ET, et al. Analytical sensitivity and clinical sensitivity of the three rapid antigen detection kits for detection of SARS-CoV-2 virus. J Clin Virol. 2020;133:104684. pmid:33176236
  275. 275. Mak GCK, Cheng PKC, Lau SSY, Wong KKY, Lau CS, Lam ETK, et al. Evaluation of rapid antigen test for detection of SARS-CoV-2 virus. J Clin Virol. 2020;129:104500. pmid:32585619
  276. 276. Merino-Amador P, Guinea J, Muñoz-Gallego I, González-Donapetry P, Galán J-C, Antona N, et al. Multicenter evaluation of the Panbio COVID-19 Rapid Antigen-Detection Test for the diagnosis of SARS-CoV-2 infection. Clin Microbiol Infect. 2020;27(5):758–61. pmid:33601009
  277. 277. Mertens P, De Vos N, Martiny D, Jassoy C, Mirazimi A, Cuypers L, et al. Development and potential usefulness of the COVID-19 Ag Respi-Strip diagnostic assay in a pandemic context. Front Med. 2020;7:225. pmid:32574326
  278. 278. Nalumansi A, Lutalo T, Kayiwa J, Watera C, Balinandi S, Kiconco J, et al. Field evaluation of the performance of a SARS-CoV-2 antigen rapid diagnostic test in Uganda using nasopharyngeal samples. Int J Infect Dis. 2020;104:282–6. pmid:33130198
  279. 279. Olearo F, Nörz D, Heinrich F, Sutter JP, Rödel K, Schultze A, et al. Handling and accuracy of four rapid antigen tests for the diagnosis of SARS-CoV-2 compared to RT-qPCR. J Clin Virol. 2020;137:104782. pmid:33711691
  280. 280. Parada-Ricart E, Gomez-Bertomeu F, Picó-Plana E, Olona-Cabases M. Usefulness of the antigen for diagnosing SARS-CoV-2 infection in patients with and without symptoms. Enferm Infecc Microbiol Clin (Engl Ed). 2020 Oct 1. pmid:33127162
  281. 281. Pekosz A, Cooper C, Parvu V, Li M, Andrews J, Manabe YCC, et al. Antigen-based testing but not real-time PCR correlates with SARS-CoV-2 virus culture. Clin Infect Dis. 2020 Jan 20. pmid:33479756
  282. 282. Perchetti GA, Huang ML, Mills MG, Jerome KR, Greninger AL. Analytical sensitivity of the Abbott BinaxNOW COVID-19 Ag CARD. J Clin Microbiol. 2021;59(3):e02880–20. pmid:33310764
  283. 283. Pilarowski G, Lebel P, Sunshine S, Liu J, Crawford E, Marquez C, et al. Performance characteristics of a rapid SARS-CoV-2 antigen detection assay at a public plaza testing site in San Francisco. J Infect Dis. 2020;223(7):1139–44. pmid:33173911
  284. 284. Porte L, Legarraga P, Iruretagoyena M, Vollrath V, Pizarro G, Munita J, et al. Rapid SARS-CoV-2 antigen detection by immunofluorescence—a new tool to detect infectivity. medRxiv. 2020 Oct 6.
  285. 285. Porte L, Legarraga P, Vollrath V, Aguilera X, Munita JM, Araos R, et al. Evaluation of a novel antigen-based rapid detection test for the diagnosis of SARS-CoV-2 in respiratory samples. Int J Infect Dis. 2020;99:328–33. pmid:32497809
  286. 286. Schildgen V, Demuth S, Lüsebrink J, Schildgen O. Limits and opportunities of SARS-CoV-2 antigen rapid tests—an experience based perspective. Pathogens. 2020;10(1):38. pmid:33466537
  287. 287. Schwob J-M, Miauton A, Petrovic D, Perdrix J, Senn N, Jaton K, et al. Antigen rapid tests, nasopharyngeal PCR and saliva PCR to detect SARS-CoV-2: a prospective comparative clinical trial. medRxiv. 2020 Nov 24.
  288. 288. Scohy A, Anantharajah A, Bodeus M, Kabamba-Mukadi B, Verroken A, Rodriguez-Villalobos H. Low performance of rapid antigen detection test as frontline testing for COVID-19 diagnosis. J Clin Virol. 2020;129:104455. pmid:32485618
  289. 289. Shrestha B, Neupane A, Pant S, Shrestha A, Bastola A, Rajbhandari B, et al. Sensitivity and specificity of lateral flow antigen test kits for COVID-19 in asymptomatic population of quarantine centre of province 3. Kathmandu Univ Med J (KUMJ). 2020;18(2):36–9. pmid:33605236
  290. 290. Takeda Y, Mori M, Omi K. SARS-CoV-2 qRT-PCR Ct value distribution in Japan and possible utility of rapid antigen testing kit. medRxiv. 2020 Jun 19.
  291. 291. Toptan T, Eckermann L, Pfeiffer A, Hoehl S, Ciesek S, Drosten C, et al. Evaluation of a SARS-CoV-2 rapid antigen test: potential to help reduce community spread? J Clin Virol. 2020;135:104713. pmid:33352470
  292. 292. Torres I, Poujois S, Albert E, Colomina J, Navarro D. Real-life evaluation of a rapid antigen test (Panbio COVID-19 Ag Rapid Test Device) for SARS-CoV-2 detection in asymptomatic close contacts of COVID-19 patients. Clin Microbiol Infect. 2020;27(4):636.E1–4. pmid:33421573
  293. 293. Van der Moeren N, Zwart V, Lodder E, Van den Bijllaardt W, Van Esch H, Stohr J, et al. Performance evaluation of a SARS-CoV-2 rapid antigentest: test performance in the community in the Netherlands. medRxiv. 2020 Oct 21.
  294. 294. Veyrenche N, Bollore K, Pisoni A, Bedin AS, Mondain AM, Ducos J, et al. Diagnosis value of SARS-CoV-2 antigen/antibody combined testing using rapid diagnostic tests at hospital admission. J Med Virol. 2020;39(5):3069–76. pmid:33554363
  295. 295. Weitzel T, Legarraga P, Iruretagoyena M, Pizarro G, Vollrath V, Araos R, et al. Comparative evaluation of four rapid SARS-CoV-2 antigen detection tests using universal transport medium. Travel Med Infect Dis. 2020;39:101942. pmid:33278609
  296. 296. Young S, Taylor SN, Cammarata CL, Varnado KG, Roger-Dalbert C, Montano A, et al. Clinical evaluation of BD Veritor SARS-CoV-2 point-of-care test performance compared to PCR-based testing and versus the Sofia 2 SARS Antigen point-of-care test. J Clin Microbiol. 2020;59(1):e02338–20. pmid:33023911
  297. 297. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71. pmid:33782057
  298. 298. Kissler SM, Fauver JR, Mack C, Olesen SW, Tai C, Shiue KY, et al. SARS-CoV-2 viral dynamics in acute infections. medRxiv. 2020 Dec 2.
  299. 299. Lee S, Kim T, Lee E, Lee C, Kim H, Rhee H, et al. Clinical course and molecular viral shedding among asymptomatic and symptomatic patients with SARS-CoV-2 infection in a community treatment center in the Republic of Korea. JAMA Intern Med. 2020;180(11):1447–52. pmid:32780793
  300. 300. Kociolek LK, Muller WJ, Yee R, Dien Bard J, Brown CA, Revell PA, et al. Comparison of upper respiratory viral load distributions in asymptomatic and symptomatic children diagnosed with SARS-CoV-2 infection in pediatric hospital testing programs. J Clin Microbiol. 2020;59(1):e02593–20. pmid:33093026
  301. 301. Turner F, Vandenberg A, Slepnev VI, Car S, Starritt RE, Seger MV, et al. Post-disease divergence in SARS-CoV-2 RNA detection between nasopharyngeal, anterior nares and saliva/oral fluid specimens—significant implications for policy & public health. medRxiv. 2021 Jan 26.
  302. 302. Bossuyt PM, Reitsma JB, Bruns DE, Gatsonis CA, Glasziou PP, Irwig L, et al. STARD 2015: an updated list of essential items for reporting diagnostic accuracy studies. BMJ. 2015;351:h5527. pmid:26511519
  303. 303. Paul Ehrlich Institute. Comparative evaluation of the sensitivities of SARSCoV-2 antigen rapid tests Langen: Federal Institute for Vaccines and Biomedicines; 2020.