Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
ACS Appl Mater Interfaces ; 15(23): 27612-27623, 2023 Jun 14.
Article in English | MEDLINE | ID: covidwho-20243632

ABSTRACT

The extensive research into developing novel strategies for detecting respiratory syndrome coronavirus 2 (SARS-CoV-2) antigens in clinical specimens, especially the sensitive point-of-care testing method, is still urgently needed to reach rapid screening of viral infections. Herein, a new lateral flow immunoassay (LFIA) platform was reported for the detection of SARS-CoV-2 spike-S1 protein antigens, in which four sensitive and specific SARS-CoV-2 mouse monoclonal antibodies (MmAbs) were tailored by using quantum dot (QD)-loaded dendritic mesoporous silica nanoparticles modified further for achieving the -COOH group surface coating (named Q/S-COOH nanospheres). Importantly, compact QD adsorption was achieved in mesoporous channels of silica nanoparticles on account of highly accessible central-radial pores and electrostatic interactions, leading to significant signal amplification. As such, a limit of detection for SARS-CoV-2 spike-S1 testing was found to be 0.03 ng/mL, which is lower compared with those of AuNPs-LFIA (traditional colloidal gold nanoparticles, Au NPs) and enzyme-linked immunosorbent assay methods. These results show that optimizing the affinity of antibody and the intensity of fluorescent nanospheres simultaneously is of great significance to improve the sensitivity of LFIA.


Subject(s)
COVID-19 , Metal Nanoparticles , Nanospheres , Animals , Mice , SARS-CoV-2 , COVID-19/diagnosis , Gold , Silicon Dioxide , Immunoassay/methods , Antibodies, Viral , Sensitivity and Specificity
2.
Int J Mol Sci ; 24(11)2023 May 31.
Article in English | MEDLINE | ID: covidwho-20237927

ABSTRACT

Rapid, accurate, and convenient diagnosis is essential for effective disease management. Various detection methods, such as enzyme-linked immunosorbent assay, have been extensively used, with lateral flow immunoassay (LFIA) recently emerging as a major diagnostic tool. Nanoparticles (NPs) with characteristic optical properties are used as probes for LFIA, and researchers have presented various types of optical NPs with modified optical properties. Herein, we review the literature on LFIA with optical NPs for the detection of specific targets in the context of diagnostics.


Subject(s)
Metal Nanoparticles , Nanoparticles , Immunoassay/methods , Enzyme-Linked Immunosorbent Assay , Gold , Limit of Detection
3.
J Chromatogr A ; 1704: 464109, 2023 Aug 16.
Article in English | MEDLINE | ID: covidwho-20230627

ABSTRACT

The shift from testing at centralized diagnostic laboratories to remote locations is being driven by the development of point-of-care (POC) instruments and represents a transformative moment in medicine. POC instruments address the need for rapid results that can inform faster therapeutic decisions and interventions. These instruments are especially valuable in the field, such as in an ambulance, or in remote and rural locations. The development of telehealth, enabled by advancements in digital technologies like smartphones and cloud computing, is also aiding in this evolution, allowing medical professionals to provide care remotely, potentially reducing healthcare costs and improving patient longevity. One notable POC device is the lateral flow immunoassay (LFIA), which played a major role in addressing the COVID-19 pandemic due to its ease of use, rapid analysis time, and low cost. However, LFIA tests exhibit relatively low analytical sensitivity and provide semi-quantitative information, indicating either a positive, negative, or inconclusive result, which can be attributed to its one-dimensional format. Immunoaffinity capillary electrophoresis (IACE), on the other hand, offers a two-dimensional format that includes an affinity-capture step of one or more matrix constituents followed by release and electrophoretic separation. The method provides greater analytical sensitivity, and quantitative information, thereby reducing the rate of false positives, false negatives, and inconclusive results. Combining LFIA and IACE technologies can thus provide an effective and economical solution for screening, confirming results, and monitoring patient progress, representing a key strategy in advancing diagnostics in healthcare.


Subject(s)
COVID-19 , Pandemics , Humans , COVID-19/diagnosis , Laboratories , Smartphone , Immunoassay/methods , COVID-19 Testing
4.
Sens Actuators B Chem ; 389: 133898, 2023 Aug 15.
Article in English | MEDLINE | ID: covidwho-2318742

ABSTRACT

Equipment-free colorimetric-based lateral flow immunoassay (LFIA) is the most convenient and popular tool for various applications, including diagnostic tools requiring high sensitivity for the detection of pathogens. Thus, improvements and developments of LFIA are constantly being reported. Herein, we enriched the sensitivity of LFIA using the gold enhancement principle, emphasizing needlessly complicated apparatus, only one step for the strip test operation, and typical time incubation (15 min) process. Self-enhanced LFIA was then executed for subsequent flows by overlapping the additionally enhanced pad composed of gold ions and reducing agent on the conjugate pad and the sample pad. Self-enhanced LFIA was performed to detect SARS-CoV-2 antigens in saliva. The obtained result depicted that the achieved sensitivity was up to tenfold compared with that of conventional LFIA by visual measurements. The detection limits of self-enhanced LFIA detecting nucleocapsid protein antigens in the saliva sample was 0.50 and 0.10 ng/mL employed by naked eye detection and calibration curve-based calculation, respectively. When the proposed device was applied to 207 human saliva samples, the diagnostic performance presented a 96.10 % sensitivity and 99.23 % specificity. This self-enhanced LFIA could be implemented in large-scale production and demonstrates higher sensitivity with effortless use, which meets the requirements for point-of-care testing and on-field mass screening.

5.
Sensors and Actuators B: Chemical ; 389, 2023.
Article in English | Scopus | ID: covidwho-2298821

ABSTRACT

Lateral flow immunoassay (LFIA) is one of the most common analytical platforms for point-of-care testing (POCT), which is capable of large-scale primary screening and home self-testing of infectious diseases. However, the sensitivity of conventional AuNPs-based LFIA is relatively low and more prone to false negatives. Herein, we report a novel LFIA based on gold-core-silver-shell bimetallic nanoparticles (Au4-ATP@Ag NPs) emitting Surface-enhanced Raman scatting (SERS) and Photothermal (PT) effect, named SERS/PT-based dual-modal LFIA (SERS/PT-dmLFIA), for the antigen detection of infectious diseases pathogens, which displayed an excellent performance. For influenza A virus (IAV), influenza B virus (IBV), and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) N protein detection, the limit of detections (LoD) with Raman as signal were 31.25, 93.75, and 31.25 pg mL-1 respectively, and the LoDs with temperature difference (∆T) as signal were as low as 15.63, 187.5, and 15.63 pg mL-1 respectively, which were over 4-fold more sensitive than visual-based LFIA. The proposed SERS/PT-dmLFIA was used for detecting virus antigen in pharyngeal swabs and showed ideal coincidence rate of over 95% compared to the commercialized assays. In addition, we explored the development of multiplex SERS/PT-dmLFIA that can detect IAV, IBV, and SARS-CoV-2 antigens simultaneously without cross reactivity. Overall, the SERS/PT-dmLFIA for antigen detection not only exhibits high sensitivity, accuracy and specificity, but also have characteristics of rapidity and simplicity, which holds high potential for rapid diagnosis of infectious diseases in laboratory testing, mass screening, and home self-testing. © 2023 Elsevier B.V.

6.
AIMS Microbiol ; 9(2): 375-401, 2023.
Article in English | MEDLINE | ID: covidwho-2304098

ABSTRACT

Over the last three years, after the outbreak of the COVID-19 pandemic, an unprecedented number of novel diagnostic tests have been developed. Assays to evaluate the immune response to SARS-CoV-2 have been widely considered as part of the control strategy. The lateral flow immunoassay (LFIA), to detect both IgM and IgG against SARS-CoV-2, has been widely studied as a point-of-care (POC) test. Compared to laboratory tests, LFIAs are faster, cheaper and user-friendly, thus available also in areas with low economic resources. Soon after the onset of the pandemic, numerous kits for rapid antibody detection were put on the market with an emergency use authorization. However, since then, scientists have tried to better define the accuracy of these tests and their usefulness in different contexts. In fact, while during the first phase of the pandemic LFIAs for antibody detection were auxiliary to molecular tests for the diagnosis of COVID-19, successively these tests became a tool of seroprevalence surveillance to address infection control policies. When in 2021 a massive vaccination campaign was implemented worldwide, the interest in LFIA reemerged due to the need to establish the extent and the longevity of immunization in the vaccinated population and to establish priorities to guide health policies in low-income countries with limited access to vaccines. Here, we summarize the accuracy, the advantages and limits of LFIAs as POC tests for antibody detection, highlighting the efforts that have been made to improve this technology over the last few years.

7.
Anal Chim Acta ; 1255: 341102, 2023 May 15.
Article in English | MEDLINE | ID: covidwho-2288795

ABSTRACT

Although many approaches have been developed for the quick assessment of SARS-CoV-2 infection, few of them are devoted to the detection of the neutralizing antibody, which is essential for assessing the effectiveness of vaccines. Herein, we developed a tri-mode lateral flow immunoassay (LFIA) platform based on gold-silver alloy hollow nanoshells (Au-Ag HNSs) for the sensitive and accurate quantification of neutralizing antibodies. By tuning the shell-to-core ratio, the surface plasmon resonance (SPR) absorption band of the Au-Ag HNSs is located within the near infrared (NIR) region, endowing them with an excellent photothermal effect under the irradiation of optical maser at 808 nm. Further, the Raman reporter molecule 4-mercaptobenzoic acid (MBA) was immobilized on the gold-silver alloy nanoshell to obtain an enhanced SERS signal. Thus, these Au-Ag HNSs could provide colorimetric, photothermal and SERS signals, with which, tri-mode strips for SARS-CoV-2 neutralizing antibody detection were constructed by competitive immunoassay. Since these three kinds of signals could complement one another, a more accurate detection was achieved. The tri-mode LFIA achieved a quantitative detection with detection limit of 20 ng/mL. Moreover, it also successfully detected the serum samples from 98 vaccinated volunteers with 79 positive results, exhibiting great application value in neutralizing antibody detection.


Subject(s)
Antibodies, Neutralizing , COVID-19 , Immunoassay , Nanoshells , SARS-CoV-2 , Spectrum Analysis, Raman , Humans , Alloys , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/immunology , Colorimetry/methods , COVID-19/diagnosis , COVID-19/immunology , Gold , Immunoassay/instrumentation , Immunoassay/methods , Metal Nanoparticles , SARS-CoV-2/immunology , Silver , Spectrum Analysis, Raman/methods
8.
ACS Appl Mater Interfaces ; 2023 Feb 13.
Article in English | MEDLINE | ID: covidwho-2262158

ABSTRACT

Developing a rapid antibody-based detection method is of great importance for preventing and controlling the spread of coronavirus disease 2019 (COVID-19). Among the antibody-based methods for point-of-care (POC) detection, lateral flow immunoassay (LFIA) is the most widely used. However, LFIA still has the disadvantage of low sensitivity. In this work, an ReSe2 nanosheet with a thickness of 10-20 nm was prepared by liquid exfoliation and applied as the label in a photothermal LFIA due to its high photothermal conversion efficiency and high photothermal stability. An integrated detection device was introduced for rapid, on-site, and highly sensitive assay of the human antisevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Spike (S) protein IgG antibodies. The device mainly included a rhenium diselenide (ReSe2) nanosheet-based photothermal LFIA, a portable laser, and a smartphone with a portable thermal imager, which was used to record and analyze the thermal signal of the LFIA test zone. The human anti-SARS-COV-2 S protein IgG antibodies in buffer solution can be detected in a portable box within 10 min, with a thermal signal detection limit of 0.86 ng mL-1, which was 108-fold lower than that of the colorimetric signal. The integrated device can detect values as low as 2.76 ng mL-1 of the human anti-SARS-COV-2 S protein IgG antibodies in 50% serum. The integrated device showed great potential for rapid and home self-testing diagnosis of COVID-19.

9.
Front Immunol ; 14: 1111644, 2023.
Article in English | MEDLINE | ID: covidwho-2272512

ABSTRACT

Development and validation of rapid and easy-to-perform diagnostics continue to be a high priority during the current COVID-19 pandemic. Although vaccines are now widely available, early detection and consistent transmission control provide ideal means to mitigate the spread of SARS-CoV-2. Nucleic acid-based real-time PCR tests are widely acknowledged as the gold standard for reliable diagnosis of COVID-19 infection. These tests are based on detecting viable or nonviable viral nucleic acids. SARS-CoV-2 spike protein is an alternative and ideal target for SARS-CoV-2 diagnosis in the early phase of infection, but point-of-care kits to detect the SARS-CoV-2 spike protein are limited. Here we describe a rapid and convenient method based on Lateral Flow Immunoassay (LFIA) to detect SARS-CoV-2 spike proteins, including SARS-CoV-2 variants (A.23.1, B.1.1.1, 1.617.2, B.1.1.7, B.1.351, P.1, N501Y, R.1, P681H, P3, UK, and South African) within 5 to 10 minutes. We generated highly specific monoclonal antibodies (mAbs) against rationally designed SARS-CoV-2 spike protein. Matched pair mAbs were selected by epitope mapping and employed as antigen capture reagents by spotting onto a nitrocellulose membrane and as detector reagents by conjugation with colloidal gold nanoparticles. We evaluated the performance of the LFIA using recombinant spike proteins of SARS-CoV-2 and several SARS-CoV-2 variants. The specificity of the LFIA was assessed using heat-inactivated SARS-CoV-2 and related human coronaviruses (HCoV-OC43, HCoV-229E, HCoV-HKU1, and HCoV-NL63) and an FDA-approved respiratory pathogens (RP) panel. The assay exhibited 98% specificity and acceptable performance with respect to the minimum limit of detection (25 ng/test) in validation tests. This new LFIA provides improved performance for the early diagnosis of SARS-CoV-2, particularly for home monitoring and in situations with limited access to molecular methods.


Subject(s)
COVID-19 , Metal Nanoparticles , Humans , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/analysis , COVID-19 Testing , Point-of-Care Systems , Pandemics , Gold , Sensitivity and Specificity , Immunoassay/methods
10.
Biochim Biophys Acta Gen Subj ; 1867(1): 130266, 2023 01.
Article in English | MEDLINE | ID: covidwho-2253544

ABSTRACT

BACKGROUND: Rapid antigen assays have been attractive for decentralized, point of care diagnostics because of their low cost, robustness, and ease of use. The development of a diagnostic assay for a newly emerging infectious disease needs to take into account the progression of a disease, whether there is human to human transmission, and patient biomarker levels with time, and these all impact the choice of antigen targets and affinity agents. SCOPE OF REVIEW: The factors involved in the biophysical design of rapid antigen immunoassays are discussed, focusing on antigen selection and designing for cross-reactivity. State of the art in the biophysical characterization of protein-ligand or antigen-antibody interactions, the different types of affinity agents used in immunoassays, and biochemical conjugation strategies are described. MAJOR CONCLUSIONS: Antigen choice is a critical factor in immunoassay diagnostic development, and should account for the properties of the virion, virus, and disease progression. Biophysical and biochemical aspects of immunoassays are critical for performance. GENERAL SIGNIFICANCE: This review can serve as an instructive guide to aid in diagnostic development for future emerging diseases.


Subject(s)
Proteins , Humans , Immunoassay , Biomarkers
11.
Nano Res ; : 1-11, 2022 Oct 22.
Article in English | MEDLINE | ID: covidwho-2286179

ABSTRACT

A lateral flow immunoassay (LFA) biosensor that allows the sensitive and accurate identification of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other common respiratory viruses remains highly desired in the face of the coronavirus disease 2019 pandemic. Here, we propose a multiplex LFA method for the on-site, rapid, and highly sensitive screening of multiple respiratory viruses, using a multilayered film-like fluorescent tag as the performance enhancement and signal amplification tool. This film-like three-dimensional (3D) tag was prepared through the layer-by-layer assembly of highly photostable CdSe@ZnS-COOH quantum dots (QDs) onto the surfaces of monolayer graphene oxide nanosheets, which can provide larger reaction interfaces and specific active surface areas, higher QD loads, and better luminescence and dispersibility than traditional spherical fluorescent microspheres for LFA applications. The constructed fluorescent LFA biosensor can simultaneously and sensitively quantify SARS-CoV-2, influenza A virus, and human adenovirus with low detection limits (8 pg/mL, 488 copies/mL, and 471 copies/mL), short assay time (15 min), good reproducibility, and high accuracy. Moreover, our proposed assay has great potential for the early diagnosis of respiratory virus infections given its robustness when validated in real saliva samples. Electronic Supplementary Material: Supplementary material (Section S1 Experimental section, Section S2 Calculation of the maximum number of QDs on the GO@TQD nanofilm, Section S3 Optimization of the LFA method, and Figs. S1-S17 mentioned in the main text) is available in the online version of this article at 10.1007/s12274-022-5043-6.

12.
Lab Med ; 2022 Jul 27.
Article in English | MEDLINE | ID: covidwho-2268872

ABSTRACT

Coronavirus disease 2019 is a serious threat to human life, and early diagnosis and screening can help control the COVID-19 pandemic. The high sensitivity of reverse transcriptase-polymerase chain reaction (RT-PCR) assay is the gold standard for the diagnosis of COVID-19, but there are still some false-negative results. Rapid antigen detection (RAD) is recommended by the World Health Organization (WHO) as a screening method for COVID-19. This review analyzed the characteristics of RDT and found that although the overall sensitivity of RAD was not as high as that of RT-PCR, but RAD was more sensitive in COVID-19 patients within 5 days of the onset of symptoms and in COVID-19 patients with Ct ≤ 25. Therefore, RAD can be used as an adjunct to RT-PCR for screening patients with early COVID-19. Finally, this review provides a combined diagnostic protocol for RAD and nucleic acid testing with the aim of providing a feasible approach for COVID-19 screening.

13.
Clin Infect Dis ; 2022 Aug 01.
Article in English | MEDLINE | ID: covidwho-2250471

ABSTRACT

BACKGROUND: We explore SARS-CoV-2 antibody lateral flow immunoassay (LFIA) performance under field conditions compared to laboratory-based electrochemiluminescence immunoassay (ECLIA) and live virus neutralisation. METHODS: In July 2021, 3758 participants performed, at home, a self-administered Fortress LFIA on finger-prick blood, reported and submitted a photograph of the result, and provided a self-collected capillary blood sample for assessment of IgG antibodies using the Roche Elecsys® Anti-SARS-CoV-2 ECLIA. We compared the self-reported LFIA result to the quantitative ECLIA and checked the reading of the LFIA result with an automated image analysis (ALFA). In a subsample of 250 participants, we compared the results to live virus neutralisation. RESULTS: Almost all participants (3593/3758, 95.6%) had been vaccinated or reported prior infection. Overall, 2777/3758 (73.9%) were positive on self-reported LFIA, 2811/3457 (81.3%) positive by LFIA when ALFA-reported, and 3622/3758 (96.4%) positive on ECLIA (using the manufacturer reference standard threshold for positivity of 0.8 U ml-1). Live virus neutralisation was detected in 169 of 250 randomly selected samples (67.6%); 133/169 were positive with self-reported LFIA (sensitivity 78.7%; 95% CI 71.8, 84.6), 142/155 (91.6%; 86.1, 95.5) with ALFA, and 169 (100%; 97.8, 100.0) with ECLIA. There were 81 samples with no detectable virus neutralisation; 47/81 were negative with self-reported LFIA (specificity 58.0%; 95% CI 46.5, 68.9), 34/75 (45.3%; 33.8, 57.3) with ALFA, and 0/81 (0%; 0.0, 4.5) with ECLIA. CONCLUSIONS: Self-administered LFIA is less sensitive than a quantitative antibody test, but the positivity in LFIA correlates better than the quantitative ECLIA with virus neutralisation.

14.
Biosensors (Basel) ; 13(3)2023 Mar 06.
Article in English | MEDLINE | ID: covidwho-2269454

ABSTRACT

Currently, the global trend of several hundred thousand new confirmed COVID-19 patients per day has not abated significantly. Serological antibody detection has become an important tool for the self-screening of people. While the most commonly used colorimetric lateral flow immunoassay (LFIA) methods for the detection of COVID-19 antibodies are limited by low sensitivity and a lack of quantification ability. This leads to poor accuracy in the screening of early COVID-19 patients. Therefore, it is necessary to develop an accurate and sensitive autonomous antibody detection technique that will effectively reduce the COVID-19 infection rate. Here, we developed a three-line LFIA immunoassay based on polydopamine (PDA) nanoparticles for COVID-19 IgG and IgM antibodies detection to determine the degree of infection. The PDA-based three-line LFIA has a detection limit of 1.51 and 2.34 ng/mL for IgM and IgG, respectively. This assay reveals a good linearity for both IgM and IgG antibodies detection and is also able to achieve quantitative detection by measuring the optical density of test lines. In comparison, the commercial AuNP-based LFIA showed worse quantification results than the developed PDA-based LFIA for low-concentration COVID-19 antibody samples, making it difficult to distinguish between negative and positive samples. Therefore, the developed PDA-based three-line LFIA platform has the accurate quantitative capability and high sensitivity, which could be a powerful tool for the large-scale self-screening of people.


Subject(s)
COVID-19 , Metal Nanoparticles , Nanoparticles , Humans , COVID-19/diagnosis , Immunoassay/methods , Immunoglobulin M , Immunoglobulin G
15.
Nanomedicine ; 47: 102624, 2022 Nov 01.
Article in English | MEDLINE | ID: covidwho-2242819

ABSTRACT

Respiratory viruses usually induced similar clinical symptoms at early infection. Herein, we presented a multichannel surface-enhanced Raman scattering-based lateral flow immunoassay (SERS-based LFA) using high-performance magnetic SERS tags for the simultaneous ultrasensitive detection of respiratory viruses, namely influenza A virus (H1N1), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and respiratory syncytial virus (RSV) in biological samples. As-prepared magnetic SERS tags can directly enrich and capture target viruses without pretreatment of samples, avoiding the interference of impurities in the samples as well as improving the sensitivity. With the capture-detection method, the detection limits of the proposed assay reached 85 copies mL-1, 8 pg mL-1, and 8 pg mL-1 for H1N1, SARS-CoV-2 and RSV, respectively. Moreover, the detection properties of the proposed method for target viruses in throat swab samples were verified, suggesting its remarkable potential for the early and rapid differential diagnosis of respiratory viruses.

16.
Biosensors and Bioelectronics: X ; : 100316.0, 2023.
Article in English | ScienceDirect | ID: covidwho-2228826

ABSTRACT

Many bacteria and viruses are spreading in the air in the living environment, and high concentrations of viruses entering the human body will cause harm. This research is committed to developing a virus collector, which is used to collect influenza and coronavirus in the air. In our system, the intake fan can beget negative pressure into the water circulation channel and bring the virus into it, and then the sensor chip will obtain the electrical signal. In this study, we successfully used methylene blue to simulate viruses in the air. The result of this experiment showed that the distance between the air virus collector and the atomizer was 30 cm in height, 60 cm & 90 cm in length. The capture efficiency was respectively 1.1% and 0.8%. Also, we use lateral flow immunochromatographic assay to detect the collected samples of Influenza H1N1 Hemagglutinin Protein, and the actual limit of detection is 16.97 ng/ml. In addition, the experiments also proved that the water circulation device in this study could accumulate the methylene blue samples onto the sensor. In the future, it can be used with the sensor chip as a virus detection platform which can collect and monitor viruses simultaneously. By using this device, people can be warned and take the necessary precautions to reduce the chance of the transmission of viruses.

17.
Front Bioeng Biotechnol ; 10: 1090281, 2022.
Article in English | MEDLINE | ID: covidwho-2230861

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is still in an epidemic situation, which poses a serious threat to the safety of people and property. Rapid diagnosis and isolation of infected individuals are one of the important methods to control virus transmission. Existing lateral flow immunoassay techniques have the advantages of rapid, sensitive, and easy operation, and some new options have emerged with the continuous development of nanotechnology. Such as lateral flow immunoassay test strips based on colorimetric-fluorescent dual-mode and gold nanoparticles, Surface Enhanced Raman Scattering, etc., these technologies have played an important role in the rapid diagnosis of COVID-19. In this paper, we summarize the current research progress of lateral flow immunoassay in the field of Severe Acute Respiratory Syndrome Coronavirus 2 infection diagnosis, analyze the performance of Severe Acute Respiratory Syndrome Coronavirus 2 lateral flow immunoassay products, review the advantages and limitations of different detection methods and markers, and then explore the competitive CRISPR-based nucleic acid chromatography detection method. This method combines the advantages of gene editing and lateral flow immunoassay and can achieve rapid and highly sensitive lateral flow immunoassay detection of target nucleic acids, which is expected to be the most representative method for community and clinical point-of-care testing. We hope that researchers will be inspired by this review and strive to solve the problems in the design of highly sensitive targets, the selection of detection methods, and the enhancement of CRISPR technology, to truly achieve rapid, sensitive, convenient, and specific detection of novel coronaviruses, thus promoting the development of novel coronavirus diagnosis and contributing our modest contribution to the world's fight against epidemics.

18.
Viruses ; 14(12)2022 11 23.
Article in English | MEDLINE | ID: covidwho-2216889

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus responsible for the COVID-19 pandemic. From the onset of the pandemic, rapid antigen tests have quickly proved themselves to be an accurate and accessible diagnostic platform. The initial (and still most commonly used antigen tests) for COVID-19 diagnosis were constructed using monoclonal antibodies (mAbs) specific to severe acute respiratory syndrome coronavirus (SARS-CoV) nucleocapsid protein (NP). These mAbs are able to bind SARS-CoV-2 NP due to high homology between the two viruses. However, since first being identified in 2019, SARS-CoV-2 has continuously mutated, and a multitude of variants have appeared. These mutations have an elevated risk of leading to possible diagnostic escape when using tests produced with SARS-CoV-derived mAbs. Here, we established a library of 18 mAbs specific to SARS-CoV-2 NP and used two of these mAbs (1CV7 and 1CV14) to generate a prototype antigen-detection lateral flow immunoassay (LFI). A side-by-side analysis of the 1CV7/1CV14 LFI and the commercially available BinaxNOWTM COVID-19 Antigen CARD was performed. Results indicated the 1CV7/1CV14 LFI outperformed the BinaxNOWTM test in the detection of BA.2, BA.2.12.1, and BA.5 Omicron sub-variants when testing remnant RT-PCR positive patient nasopharyngeal swabs diluted in viral transport media.


Subject(s)
COVID-19 , Humans , COVID-19/diagnosis , SARS-CoV-2/genetics , COVID-19 Testing , Pandemics , Sensitivity and Specificity , Immunoassay/methods , Antigens , Antibodies, Monoclonal
19.
Biomater Transl ; 2(1): 43-49, 2021.
Article in English | MEDLINE | ID: covidwho-2201125

ABSTRACT

The outbreak of the novel coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), spread rapidly causing a severe global health burden. The standard COVID-19 diagnosis relies heavily on molecular tests to detect viral RNA in patient samples; however, this method is costly, requires highly-equipped laboratories, multiple reagents, skilled laboratory technicians, and takes 3-6 hours to complete. To overcome these limitations, we developed a plant-based production platform for the SARS-CoV-2 receptor-binding domain as an economical source of detection reagents for a lateral-flow immunoassay strip (LFIA) which is suitable for detection of IgM/IgG antibodies in human samples. Further, we validated the plant-produced SARS-CoV-2 receptor-binding domain-based LFIA as a useful diagnostic tool for COVID-19. A total of 51 confirmed COVID-19 serum samples were tested using the LFIA, and the obtained results were consistent with those from polymerase chain reaction assays, while providing sensitivity and specificity of 94.1% and 98%, respectively. The developed LFIA is rapid, scalable, user-friendly, and relatively inexpensive with a simple test procedure, making it useful for the routine monitoring of COVID-19 in clinical settings. This study was approved on March 19, 2020 by the Ethics Committee of the Faculty of Medicine, Chulalongkorn University (COA No. 354/2020 and IRB No. 236/63).

20.
Engineering Proceedings ; 27(1), 2022.
Article in English | Scopus | ID: covidwho-2199900

ABSTRACT

Lateral flow assays (LFAs;aka. rapid tests) are inexpensive paper-based devices for rapid and specific detection of analyte of interest (e.g., COVID virus) in fluidic samples. Areas of application of LFAs cover a broad spectrum, spanning from agriculture to food/water safety to point-of-care medical testing and, most recently, to detection of COVID-19 infection. While these low-cost and rapid tests are specific to the target analyte, their sensitivity and limit of detection are far inferior to their laboratory-based counterparts. In addition, rapid tests normally cannot quantify the concentration of target analyte and only provide qualitative/binary detection. We have developed a low-cost, end-user sensing platform that significantly improves the sensitivity of rapid tests. The developed platform is based on Arduino and utilizes low-cost far infrared, single-element detectors to offer sensitive and semi-quantitative results from commercially available rapid tests. The sensing paradigm integrated to the low-cost device is based on radiometric detection of photothermal responses of rapid tests in the frequency domain when exposed to modulated laser excitation. As a proof of principle, we studied commercially available rapid tests for detection of THC (the principal psychoactive constituent of cannabis) in oral fluid with different concentrations of control positive solutions and, subsequently, interpret them with the developed sensor. Results suggest that the developed end-user sensor is not only able to improve the detection limit of the rapid test by approximately an order of magnitude from 25 ng/mL to 5 ng/mL, but also offers the ability to obtain semi-quantitative insight into concentration of THC in the fluidic samples. © 2022 by the authors.

SELECTION OF CITATIONS
SEARCH DETAIL