Skip to main content

Diagnostic utility of antigen detection rapid diagnostic tests for Covid-19: a systematic review and meta-analysis

Abstract

Background

The early detection of coronavirus disease (COVID-19) infection to improve disease management becomes the greatest challenge. Despite the high sensitivity of RT-PCR, not only it was reported that 20–67% of infected patients had false-negative results. Rapid diagnostic tests (RDTs) are widely used as a point-of-care test for SARS-CoV-2 detection in pharyngeal and blood specimens. It’s more appealing since it’s less time-consuming, doesn’t seem to be as expensive, and doesn’t need any specific training, but the poor sensitivity is the major limitation. Several reports indicated the rapid test of blood and pharyngeal samples has the same sensitivity as the RT-PCR, but some reports have lower sensitivity, especially in asymptomatic patients.

Methods

In the present survey, we investigate the eligible studies for the sensitivity and specificity of rapid tests and explore the factors that influence the result to help better diagnose COVID-19 infection. 20 studies met the inclusion criteria which imposed 33 different tests.

Results

Our findings showed the type of sample, the type of assay, the time of sampling, and the load of virus influence on the sensitivity of RDTs.

Conclusion

This research extends our knowledge of how to improve the sensitivity of RDTs to better diagnose the infected patients to address the controlling COVID-19 pandemic.

Introduction

Early coronavirus infection 2019 (COVID-19) detection is an important and challenging issue to prevent its rapid spread. Real-time PCR (RT-PCR) is a standard gold test used to detect COVID-19 in the laboratory. Despite the high sensitivity of RT-PCR, it was reported that 20–67% of infected patients have false-negative results, but RT-PCR cannot differentiate between infectious and non-infectious SARS-CoV-2 particles [1, 2]. Rapid diagnostic tests (RDTs) for SARS-CoV-2 detection in pharyngeal and blood specimens are routinely employed as point-of-care testing. Limited time and expense, as well as the need of specialized training, make these approaches more appealing, but their major limitation is their low sensitivity. It was shown that some infected individuals still have a positive RT-PCR after final recovery. This false positive can be the cause of existing RNA particles [3], while this has not been reported for RDTs. To detect the IgG and IgM Abs in the blood, plasma, and serum of patients with COVID-19, in addition to the rapid antigen (Ag) test, the rapid antibody (Ab) test is considered a timely point-of-care test [4, 5]. In a recent study, Ricks et al., comparing the two methods of RDTs and RT-PCR due to the health system cost and health impact, stated that despite the low sensitivity of RDTs to RT-PCR, it has valuable to identify the patients infected with reduced costs [6]. Besides, the ability to perform RDTs during the night, weekends, and holidays has made it a significant advantage for diagnosis in emergency medical departments.

Extensive studies have examined the usability of RDTs based on their sensitivity and specificity compared to RT-PCR. However, the reported inconsistent findings need more investigation in order to arrive at a meaningful conclusion. Several studies have shown that fast blood and throat samples had comparable sensitivities to RT-PCR, while others have found lower sensitivity, particularly in asymptomatic individuals [7,8,9,10]. These controversial results may be due to different samples and testing to various stages of infection, as many COVID-19 antigen tests are rapidly evolving and need to be systematically indicated in all of these available tests. Recent systematic reviews and Meta-analyzes of the literature have focused on the accuracy of point-of-care tests and diagnostic tests, but no detailed studies were performed on the accuracy of Ag-RDTs in the diagnosis of COVID-19 [11, 12].

In the present systematic review, we attempt to assess the diagnostic utility of antigen detection rapid diagnostic tests for COVID-19 versus RT-PCR in a different types of samples and different stages of infection determine the usability of rapid tests in the best time and sample and explore the factors that influence the result to help better diagnose COVID-19 infection.

Materials and methods

This review was performed following the PRISMA (Preferred Reporting Items for Systematic reviews and Meta-Analyses) and MOOSE (Meta-analyses Of Observational Studies in Epidemiology) guidelines [13, 14].

Search strategy

To evaluate the usability of rapid tests compared with RT-PCR, we systematically searched the electronic database, including Scopus, Medline/PubMed, EMBASE, Web of sciences (WOS), and Cochrane library using Mesh-standardized keywords: (((Rapid antigen detection test * OR RDT*) OR “Rapid Antigen Test ”[Mesh] OR “point of care testing”) AND (“Real-time PCR”[Mesh] OR “RT-PCR” OR ‎“Molecular diagnostic test” OR “RNA virus”) AND (“2019 nCoV” OR 2019nCoV OR “2019 novel coronavirus” OR “COVID 19” OR COVID19 OR “new coronavirus” OR “novel coronavirus” OR “novel coronavirus” OR “SARS CoV-2” OR (Wuhan AND (coronavirus OR “corona virus”) OR “COVID-19” OR “severe acute respiratory syndrome coronavirus 2”)) until Jan 2021. There is no restriction for time and language, and the citation lists of selected articles were hand-searched for additional papers.

Data extraction

Two reviewers independently screened the titles and abstracts of all initially found articles. Information was extracted from selected studies including the name of author, country, sample size, mean age, rapid diagnostic kits, true positive, false positive, false negative, true negative results, sensitivity, and specificity. A third reviewer was consulted to resolve any disagreements between reviewers by discussion until consensus was reached.

Eligible criteria

To understand the sensitivity and specificity of RDTs, studies that evaluated these parameters were selected. Inclusion criteria were considered as follows: the evaluation of the sensitivity and specificity of RDTs compared to the RT-PCR. All types of studies, including case/control, cohort, cross-sectional, and clinical trial studies, were included. Studies that evaluated seroprevalence, studies that investigated just cell culture assay, case reports, reviews, and studies reporting cases with incomplete information were excluded.

Statistical analysis

Cochran Chi-square test and I2 were used to assess the heterogeneity among studies. A fixed-effects model was used when I2 < 50%, while a random-effects model was selected in the case of I2 > 50%. Fixed- effect model assumes that the population effect sizes are the same for all studies [15]. In contrast, the random-effects model attempted to generalize findings beyond the included studies by assuming that the selected studies are random samples from a larger population [16]. To compare the sensitivity and specificity in RDTs compared with the RT-PCR, 95% confidence intervals (CI) were used. According to the heterogeneity test results, either Der Simonian’s and Laird’s random‑effects method or Mantel‑Haenszel’s fixed‑effects method were used to estimate the overall sensitivity and specificity and 95% confidence intervals [17]. Moreover, subgroup analysis was implemented based on the type of specimen (nasopharyngeal swab, throat washing and bronchoalveolar fluids, and Nasal sample), and symptomatic or asymptomatic patinates as an important variable which may cause heterogeneity between different samples or influence of onset of symptoms. The Egger’s test was used to investigate small study effects due to potential publication bias [18, 19]. If the findings were statistically heterogeneous, a sensitivity analysis was performed to establish the cause of heterogeneity. The randomized effects model was utilized for meta-analysis after considerable clinical heterogeneity was eliminated. P < 0.05 was considered the statistical significance (2-sided). All data were analyzed using STAT 16 (STATA Corporation, College Station, Texas).

Result

In general, 783 studies were initially collected (Fig. 1). Finally, 20 studies met our inclusion criteria, which imposed 33 different tests (including 26,056 patients, mean age range from 20.5 to 53.14 years). Eleven studies (55%) evaluated nasopharyngeal swabs [5, 6, 8, 11,12,13,14,15,16,17,18,19]. Three included studies investigated various types of samples with just one assay [20,21,22], whereas the other two articles examined one type of sample with different assays to compare the sensitivity and specificity [8, 9]. A single study performed a similar series of experiments but for rapid Abs tests with finger-stick whole-blood [10], whereas other two studies were used the same rapid Abs tests from patient’s serum [23, 24]. Characteristics of included studies are shown in Table 1.

Fig. 1
figure 1

Flow diagram of the study selection process

Table 1 Characteristics of included studies

Our pooled analysis has revealed that the sensitivity and specificity of RDTs were … and…, respectively. Some literatures analyzed the different types of specimens [25,26,27], some analyzed the sensitivity and specificity during different stages of infection [9, 25, 26, 28, 29], and some compared the diagnostic accuracy of RDTs with RT-PCR based viral load of viruses [8, 20, 28, 30,31,32,33]. The characteristics of the diagnostic values of included studies are shown in Table 2. The results of Cochrane Q and I2 statistics showed significant heterogeneity in sensitivity and specificity, so estimations of sensitivity and specificity were obtained using a random effect model. In further analysis, data were analyzed based on the type of sample and symptomatic or asymptomatic patients Table S1, S2, S3, S4, S5 and Figure S1, S2, S3, S4, S5. Summary ROC curves constructed for all assays based on Monte Carlo simulations are shown in Fig. 2. It did not apply to assessing the false results, because 7 studies (35%) did not focus on false results [8, 9, 20, 22, 24, 26, 34, 35]. Some of the current kinds of literature which focused on Ab rapid tests were excluded due to reporting the results as the separate sensitivity for IgG and IgM or analyzed data based on onset of symptoms [4, 36].

Table 2 Pooled analysis of sensitivity and specificity of included studies with 95% confidence interval
Fig. 2
figure 2

Summary receiver operating characteristic curve (ROC curve). Estimates of sensitivity and specificity for each study are

Discussion

Reverse transcription-quantitative PCR (RT-qPCR) is widely used to diagnose COVID-19. However, these tests cannot be performed in local clinics where RT-qPCR testing is unavailable, so rapid antigen tests (RATs) for COVID-19 are used for rapid diagnosis [37]. Therefore, samples should be moved to locations that have RT-qPCR capability. This process delays the test result and increases the anxiety of patients suspected of having COVID-19. To solve this problem, RATs, which do not require special and expensive machines, have been approved to detect COVID-19, and the sensitivity of these tests was compared with RT-qPCR methods [38,39,40].

The advent of rapid antigen diagnostic tests for SARS-CoV-2 has posed many challenges in terms of accessibility and performance. Further evidence on how to conduct and use such tests is needed to reach a definitive conclusion about using them. Choosing the proper virological test for COVID-19 affects early detection, rapid control, and prophylactic responses to virus outbreaks. Early detection allows promptly removing infections, thus minimizing transmission opportunities [1]. Recent interpretations have highlighted the potential application of high-frequency, low-sensitivity experiments in asymptomatic individuals. Current analysis suggests a less sensitive, but more accessible test may be preferable to detect symptomatic COVID- 19 [41]. COVID-19 Ag-RTD is a recent generation sensitive and specific antigen test for the qualitative detection of SARS-CoV-2 antigen in human nasopharyngeal swab samples. Ag-RDTs may be based on more accessible and uncomplicated samples that can even be self-collected, such as nasal swab or saliva, in addition to the nasopharyngeal swab, which needs qualified healthcare personnel and personal protective equipment to collect. Ag-RDTs directly detect SARS-CoV-2 proteins produced by virus replication in respiratory secretions [25]. Ag-RDTs, compared to methods such as nucleic acid amplification tests (NAAT) and RT-PCR, are relatively inexpensive, easy to perform, do not require infrastructure, and allow care results to be achieved in minutes. Assessments by Ricks et al. strongly supported Ag-RDT for evaluating symptomatic individuals, making it less cost-effective and practical than relying on NAT and clinical judgment [6]. They announced Ag-RDTs are more available than other methods but are usually less sensitive and specific. However, RDTs read samples with high levels of virus as positive, and even the most sensitive RDTs read samples with small amounts of the virus as negative. Ag-RDTs are estimated to be less than 80% sensitive to COVID-19, compared to > 90% for NAT [42]. Despite the lower sensitivity of Ag-RDT, it is more helpful in guiding patient management at point-of-care, repeat testing, and large-scale public health decisions on time to prevent transmission [31]. Yamayoshi et al. determined that soaking the sample immediately in the lease buffer might boost the sensitivity of RDTs after examining different samples [37]. RDTs are less sensitive for nasal examination swabs and saliva samples, according to studies examining various kinds of specimens [25]. Performing RDT with different assays is an important issue which can cause various sensitivities. A study to determine the sensitivity of varying RDT assays showed different results for positive controls in various assays that could provide appropriate methods to identify the distinct Ags and consequently test accuracy [26].

Our results showed that most of the collected samples are nasopharyngeal swabs, the study of which increases the sensitivity of RDTs results. RT-PCR analyses showed higher sensitivity in infected patients with lower Ct values. In principle, RDT susceptibility is reduced in asymptomatic patients or patients with a lower viral load. Cerutti et al., using the amount of Ct by RT-PCR, concluded that in R-Ag positive samples, Ct was significantly lower than in R-Ag negative samples. Most RT-PCR positive and R-Ag negative samples reported negative results during cell culture, which may explain the higher R-Ag sensitivity [43]. Asymptomatic persons, on the other hand, should not be ruled out, according to Pilarowski et al., since asymptomatic patients may also have a high viral load [27]. Albert et al. evaluated cell culture sensitivity with RT-PCR to identify COVID-19 in addition to RDTs. The results showed that susceptibility was reduced in cell cultures with a lower viral load, such as RDTs [31].

To assess the sensitivity of RDTs, Eshghifar et al. used different concentrations of heat-inactivated COVID-19 virus to evaluate the cut-off detection for RDTs. Their results showed that they could not determine a cut-off and reported RDTs positively only in patients with high viral load [22]. It’s impossible to rule out the possibility of interactions throughout the test. According to certain studies, testing fewer than 5 days after the beginning of symptoms improves sensitivity [9, 22, 34], which contradicts prior claims that the infectious virus load declines after 7 to 10 days [44].Depending on the age of participants, Albert et al. reported that RDTs were less sensitive in children than in adults [31].

Our analysis showed that rapid detection of Ab is less sensitive to RDT and is associated with many false positives. In their study, Pere et al. found that a recent history of infection with cold Abs led to a false positive increase in rapid Ab testing, especially for IgM, which reduced the validity of the test [10]. Faber et al. carried out a similar investigation, and their results revealed that false positives are more likely among pregnant women [23]. One of the causes for the quick Ab test’s poor accuracy might be the patients’ short-term safety [45]. Many studies focus on the accuracy of serological tests, and as expected, their findings show less accuracy in serological tests [11, 46]. Our findings showed the type of sample, type of assay, time of sampling, and the load of virus influence on the sensitivity of RDTs. Despite domestic vaccination, there are still significant new cases, especially in low- and middle-income countries. Hence, advances in rapid detection at lower costs remain a significant challenge. The findings of this meta-analysis have important implications for the development of the RDT technique in the detection of SARS-CoV-2 with the highest possible sensitivity and specificity.

Conclusions

The main goal of the current study was to determine the accuracy of RDTs. We showed the explanations for low sensitivity in RDTs such as type of specimen, the timing of sampling, type of assay, and viral load.; and by considering them, RDTs can be used to identify the suspected patients in the early stage of disease with desirable sensitivity and specificity and help control COVID-19 pandemic.

Limitations

The major limitation of this study is the accuracy of RDTs affecting the spread of the COVID-19 virus or not? In this regard, more research is required to determine the efficacy of RDTs in detecting the various type of COVID-19 viruses. Another issue that was not addressed in this study was whether false results in RDTs. This is because seven studies did not report false results.

Availability of data and materials

The datasets generated and/or analyzed during the current study are available in the [Pubmed, Web of Science, Scopus, EM Base] repository.

Abbreviations

COVID-19:

Coronavirus infection 2019

RT-PCR:

Real-time PCR

Ag:

Antigen

Ab:

Antibody

WOS:

Web of sciences

CI:

Confidence intervals

RT-qPCR:

Reverse transcription-quantitative PCR

RATs:

Rapid antigen tests

NAAT:

Nucleic acid amplification tests

References

  1. Wiersinga WJ, Rhodes A, Cheng AC, Peacock SJ, Prescott HC. Pathophysiology, transmission, diagnosis, and treatment of coronavirus disease 2019 (COVID-19): a review. JAMA. 2020;324(8):782–93.

    Article  CAS  PubMed  Google Scholar 

  2. Singanayagam A, Patel M, Charlett A, Bernal JL, Saliba V, Ellis J, et al. Duration of infectiousness and correlation with RT-PCR cycle threshold values in cases of COVID-19, England, January to May 2020. Eurosurveillance. 2020;25(32):2001483.

    Article  CAS  PubMed Central  Google Scholar 

  3. Song K-H, Kim D-M, Lee H, Ham SY, Oh S-M, Jeong H, et al. Dynamics of viral load and anti-SARS-CoV-2 antibodies in patients with positive RT-PCR results after recovery from COVID-19. Korean J Intern Med. 2021;36(1):11.

    Article  CAS  PubMed  Google Scholar 

  4. Uwamino Y, Wakui M, Aoki W, Kurafuji T, Yanagita E, Morita M, et al. Evaluation of the usability of various rapid antibody tests in the diagnostic application for COVID-19. Ann Clin Biochem. 2021;58(3):174–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Li Z, Yi Y, Luo X, Xiong N, Liu Y, Li S, et al. Development and clinical application of a rapid IgM-IgG combined antibody test for SARS‐CoV‐2 infection diagnosis. J Med Virol. 2020;92(9):1518–24.

    Article  CAS  PubMed  Google Scholar 

  6. Ricks S, Kendall EA, Dowdy DW, Sacks JA, Schumacher SG, Arinaminpathy N. Quantifying the potential value of antigen-detection rapid diagnostic tests for COVID-19: a modelling analysis. BMC Med. 2021;19(1):1–13.

    Article  Google Scholar 

  7. 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(5):2988–91.

    Article  CAS  PubMed  Google Scholar 

  8. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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.

    Article  CAS  PubMed  Google Scholar 

  10. Péré H, Bouassa R-SM, Tonen-Wolyec S, Podglajen I, Veyer D, Bélec L. Analytical performances of five SARS-CoV-2 whole-blood finger-stick IgG-IgM combined antibody rapid tests. J Virol Methods. 2021;290:114067.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Böger B, Fachi MM, Vilhena RO, Cobre AF, Tonin FS, Pontarolo R. Systematic review with meta-analysis of the accuracy of diagnostic tests for COVID-19. Am J Infect Control. 2021;49(1):21–9.

    Article  PubMed  Google Scholar 

  12. Ricco M, Ferraro P, Gualerzi G, Ranzieri S, Henry BM, Said YB, et al. Point-of-care diagnostic tests for detecting SARS-CoV-2 antibodies: a systematic review and meta-analysis of real-world data. J Clin Med. 2020;9(5):1515.

    Article  CAS  PubMed Central  Google Scholar 

  13. Stroup DF, Berlin JA, Morton SC, Olkin I, Williamson GD, Rennie D, et al. Meta-analysis of observational studies in epidemiology: a proposal for reporting. JAMA. 2000;283(15):2008–12.

    Article  CAS  PubMed  Google Scholar 

  14. Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JP, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. J Clin Epidemiol. 2009;62(10):e1–34.

    Article  PubMed  Google Scholar 

  15. Cheung MWL, Ho RC, Lim Y, Mak A. Conducting a meta-analysis: basics and good practices. Int J Rheum Dis. 2012;15(2):129–35.

    Article  PubMed  Google Scholar 

  16. Lim RB, Zhang MW, Ho R. Prevalence of all-cause mortality and suicide among bariatric surgery cohorts: a meta-analysis. Int J Environ Res Public Health. 2018;15(7):1519.

    Article  PubMed Central  Google Scholar 

  17. DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. 1986;7(3):177–88.

    Article  CAS  PubMed  Google Scholar 

  18. Mantel N, Haenszel W. Statistical aspects of the analysis of data from retrospective studies of disease. J Natl Cancer Inst. 1959;22(4):719–48.

    CAS  PubMed  Google Scholar 

  19. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ. 2003;327(7414):557–60.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Mak GC, Cheng PK, Lau SS, Wong KK, Lau C, Lam ET, et al. Evaluation of rapid antigen test for detection of SARS-CoV-2 virus. J Clin Virol. 2020;129:104500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Agulló V, Fernández-González M, de la Tabla VO, Gonzalo-Jiménez N, García JA, Masiá M, et al. Evaluation of the rapid antigen test Panbio COVID-19 in saliva and nasal swabs in a population-based point-of-care study. J Infect. 2021;82(5):186–230.

    Article  PubMed  Google Scholar 

  22. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Fabre M, Ruiz-Martinez S, Monserrat Cantera M, Cortizo Garrido A, Beunza Fabra Z, Peran M, et al. SARS-CoV-2 immunochromatographic IgM/IgG rapid test in pregnancy: A false friend? Ann Clin Biochem. 2021;58(2):149–52.

    Article  CAS  PubMed  Google Scholar 

  24. Montesinos I, Gruson D, Kabamba B, Dahma H, Van den Wijngaert S, Reza S, et al. Evaluation of two automated and three rapid lateral flow immunoassays for the detection of anti-SARS-CoV-2 antibodies. J Clin Virol. 2020;128:104413.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. 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 in a population-based point-of-care study. J Infect. 2021;82(5):186–230.

  26. Schildgen V, Demuth S, Lüsebrink J, Schildgen O. Limits and Opportunities of SARS-CoV-2 Antigen Rapid Tests: An Experienced-Based Perspective. Pathogens. 2021;10(1).

  27. Pilarowski G, Lebel P, Sunshine S, Liu J, Crawford E, Marquez C, et al. Performance Characteristics of a Rapid Severe Acute Respiratory Syndrome Coronavirus 2 Antigen Detection Assay at a Public Plaza Testing Site in San Francisco. J Infect Dis. 2021;223(7):1139–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Scohy A, Anantharajah A, Bodéus 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Abdel-Khalik J, Hearn T, Dickson AL, Crick PJ, Yutuc E, Austin-Muttitt K, et al. Bile acid biosynthesis in Smith-Lemli-Opitz syndrome bypassing cholesterol: Potential importance of pathway intermediates. J Steroid Biochem Mol Biol. 2021;206:105794.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Albert E, Torres I, Bueno F, Huntley D, Molla E, Fernández-Fuentes M, 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. 2021;27(3):472. e7-. e10.

    Article  PubMed  Google Scholar 

  32. 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. 2021;104:282–6.

    Article  CAS  PubMed  Google Scholar 

  33. 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. 2021;52(2):e46-e9.

    Article  Google Scholar 

  34. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Salvagno GL, Gianfilippi G, Bragantini D, Henry BM, Lippi G. Clinical assessment of the Roche SARS-CoV-2 rapid antigen test. Diagnosis (Berl). 2021;8(3):322–6.

    Article  CAS  Google Scholar 

  36. Prazuck T, Colin M, Giachè S, Gubavu C, Seve A, Rzepecki V, et al. Evaluation of performance of two SARS-CoV-2 Rapid IgM-IgG combined antibody tests on capillary whole blood samples from the fingertip. PLoS ONE. 2020;15(9):e0237694.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. 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. https://doi.org/10.3390/v12121420.

    Article  CAS  PubMed Central  Google Scholar 

  38. 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).

  39. Mak GC, Cheng PK, Lau SS, Wong KK, Lau CS, Lam ET, et al. Evaluation of rapid antigen test for detection of SARS-CoV-2 virus. J Clin Virol. 2020;129:104500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Khalid MF, Selvam K, Jeffry AJN, Salmi MF, Najib MA, Norhayati MN, et al. Performance of Rapid Antigen Tests for COVID-19 Diagnosis: A Systematic Review and Meta-Analysis. Diagnostics (Basel). 2022;12(1).

  41. Larremore DB, Wilder B, Lester E, Shehata S, Burke JM, Hay JA, et al. Test sensitivity is secondary to frequency and turnaround time for COVID-19 screening. Sci Adv. 2021;7(1).

  42. Birks JS. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane database of systematic reviews. 2006(1).

  43. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Cevik M, Tate M, Lloyd O, Maraolo AE, Schafers J, Ho A. SARS-CoV-2, SARS-CoV, and MERS-CoV viral load dynamics, duration of viral shedding, and infectiousness: a systematic review and meta-analysis. The lancet microbe. 2021;2(1):e13–22.

    Article  CAS  PubMed  Google Scholar 

  45. Augustine R, Das S, Hasan A, Abdul Salam S, Augustine P, Dalvi YB, et al. Rapid antibody-based COVID-19 mass surveillance: relevance, challenges, and prospects in a pandemic and post-pandemic world. J Clin Med. 2020;9(10):3372.

    Article  CAS  PubMed Central  Google Scholar 

  46. 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.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We wish to thank all our colleagues in Allied Health Sciences School, Ahvaz Jundishapur University of Medical Sciences.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

F.R conceived the manuscript and revised it. S.Gh, and N.N done the statistical analysis, wrote the manuscript, and prepared tables and figures. All authors have read and approved the manuscript.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflict of interest. All procedure performs in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or compare ethical strand.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ghasemi, S., Harmooshi, N.N. & Rahim, F. Diagnostic utility of antigen detection rapid diagnostic tests for Covid-19: a systematic review and meta-analysis. Diagn Pathol 17, 36 (2022). https://doi.org/10.1186/s13000-022-01215-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13000-022-01215-6