Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Small ; : e2302023, 2023 May 28.
Article in English | MEDLINE | ID: covidwho-20231378

ABSTRACT

Deoxyribonuclease-I (DNase-I), a representative endonuclease, is an important biomarker for the diagnosis of infectious diseases and cancer progression. However, enzymatic activity decreases rapidly ex vivo, which highlights the need for precise on-site detection of DNase-I. Here, a localized surface plasmon resonance (LSPR) biosensor that enables the simple and rapid detection of DNase-I is reported. Moreover, a novel technique named electrochemical deposition and mild thermal annealing (EDMIT) is applied to overcome signal variations. By taking advantage of the low adhesion of gold clusters on indium tin oxide substrates, both the uniformity and sphericity of gold nanoparticles are increased under mild thermal annealing conditions via coalescence and Ostwald ripening. This ultimately results in an approximately 15-fold decrease in LSPR signal variations. The linear range of the fabricated sensor is 20-1000 ng mL-1 with a limit of detection (LOD) of 127.25 pg mL-1 , as demonstrated by spectral absorbance analyses. The fabricated LSPR sensor stably measured DNase-I concentrations from samples collected from both an inflammatory bowel disease (IBD) mouse model, as well as human patients with severe COVID-19 symptoms. Therefore, the proposed LSPR sensor fabricated via the EDMIT method can be used for early diagnosis of other infectious diseases.

2.
Adv Biomark Sci Technol ; 2: 1-23, 2020.
Article in English | MEDLINE | ID: covidwho-2288563

ABSTRACT

Due to the unprecedented public health crisis caused by COVID-19, our first contribution to the newly launching journal, Advances in Biomarker Sciences and Technology, has abruptly diverted to focus on the current pandemic. As the number of new COVID-19 cases and deaths continue to rise steadily around the world, the common goal of healthcare providers, scientists, and government officials worldwide has been to identify the best way to detect the novel coronavirus, named SARS-CoV-2, and to treat the viral infection - COVID-19. Accurate detection, timely diagnosis, effective treatment, and future prevention are the vital keys to management of COVID-19, and can help curb the viral spread. Traditionally, biomarkers play a pivotal role in the early detection of disease etiology, diagnosis, treatment and prognosis. To assist myriad ongoing investigations and innovations, we developed this current article to overview known and emerging biomarkers for SARS-CoV-2 detection, COVID-19 diagnostics, treatment and prognosis, and ongoing work to identify and develop more biomarkers for new drugs and vaccines. Moreover, biomarkers of socio-psychological stress, the high-technology quest for new virtual drug screening, and digital applications are described.

3.
J Biophotonics ; : e202200277, 2022 Nov 23.
Article in English | MEDLINE | ID: covidwho-2252174

ABSTRACT

Infrared spectroscopy is a non-destructive and rapid characterization tool that can distinguish different viral proteins by spectral details. However, traditional infrared spectroscopy has insufficient absorption signal intensity contrast when measuring low-concentration samples. In this work, surface enhanced infrared absorption (SEIRA) spectroscopy is proposed by deploying a novel nanostructure array as SEIRA substrates. An array of gold dendric nanostructures are designed and fabricated with a precision resonance control to achieve surface enhancement covering a broadband molecular "finger-print" region. The spectral positions of the multiple resonances accurately correspond to the characteristic absorption peaks of the SARS-CoV-2 proteins. An approach for SARS-CoV-2 protein detection based on SEIRA spectroscopy is then proposed. A low concentration detection of 40 µg/ml diluted SARS-CoV-2 nucleocapsid protein is experimentally demonstrated and the enhancement factor (EF) achieved is in good agreement with simulation results. The SEIRA methodology based on broadband resonance nanostructure design provides a systematic approach for sensitive, non-destructive and rapid protein molecular detection, which could be extended to various kind of molecular characterization and biomedical diagnostics.

4.
Adv Mater Interfaces ; 10(1)2023 Jan 05.
Article in English | MEDLINE | ID: covidwho-2246135

ABSTRACT

A novel localized surface plasmon resonance (LSPR) system based on the coupling of gold nanomushrooms (AuNMs) and gold nanoparticles (AuNPs) is developed to enable a significant plasmonic resonant shift. The AuNP size, surface chemistry, and concentration are characterized to maximize the LSPR effect. A 31 nm redshift is achieved when the AuNMs are saturated by the AuNPs. This giant redshift also increases the full width of the spectrum and is explained by the 3D finite-difference time-domain (FDTD) calculation. In addition, this LSPR substrate is packaged in a microfluidic cell and integrated with a CRISPR-Cas13a RNA detection assay for the detection of the SARS-CoV-2 RNA targets. Once activated by the target, the AuNPs are cleaved from linker probes and randomly deposited on the AuNM substrate, demonstrating a large redshift. The novel LSPR chip using AuNP as an indicator is simple, specific, isothermal, and label-free; and thus, provides a new opportunity to achieve the next generation multiplexing and sensitive molecular diagnostic system.

5.
Biosens Bioelectron ; 220: 114861, 2023 Jan 15.
Article in English | MEDLINE | ID: covidwho-2244685

ABSTRACT

We propose a label-free biosensor based on a porous silicon resonant microcavity and localized surface plasmon resonance. The biosensor detects SARS-CoV-2 antigen based on engineered trimeric angiotensin converting enzyme-2 binding protein, which is conserved across different variants. Robotic arms run the detection process including sample loading, incubation, sensor surface rinsing, and optical measurements using a portable spectrometer. Both the biosensor and the optical measurement system are readily scalable to accommodate testing a wide range of sample numbers. The limit of detection is 100 TCID50/ml. The detection time is 5 min, and the throughput of one single robotic site is up to 384 specimens in 30 min. The measurement interface requires little training, has standard operation, and therefore is suitable for widespread use in rapid and onsite COVID-19 screening or surveillance.


Subject(s)
Biosensing Techniques , COVID-19 , Optical Devices , Humans , COVID-19/diagnosis , SARS-CoV-2 , Surface Plasmon Resonance
6.
ACS Appl Mater Interfaces ; 2022 Dec 01.
Article in English | MEDLINE | ID: covidwho-2133169

ABSTRACT

Low-cost, instrument-free colorimetric tests were developed to detect SARS-CoV-2 using plasmonic biosensors with Au nanoparticles functionalized with polyclonal antibodies (f-AuNPs). Intense color changes were noted with the naked eye owing to plasmon coupling when f-AuNPs form clusters on the virus, with high sensitivity and a detection limit of 0.28 PFU mL-1 (PFU stands for plaque-forming units) in human saliva. Plasmon coupling was corroborated with computer simulations using the finite-difference time-domain (FDTD) method. The strategies based on preparing plasmonic biosensors with f-AuNPs are robust to permit SARS-CoV-2 detection via dynamic light scattering and UV-vis spectroscopy without interference from other viruses, such as influenza and dengue viruses. The diagnosis was made with a smartphone app after processing the images collected from the smartphone camera, measuring the concentration of SARS-CoV-2. Both image processing and machine learning algorithms were found to provide COVID-19 diagnosis with 100% accuracy for saliva samples. In subsidiary experiments, we observed that the biosensor could be used to detect the virus in river waters without pretreatment. With fast responses and requiring small sample amounts (only 20 µL), these colorimetric tests can be deployed in any location within the point-of-care diagnosis paradigm for epidemiological control.

7.
ACS Appl Mater Interfaces ; 14(43): 48464-48475, 2022 Nov 02.
Article in English | MEDLINE | ID: covidwho-2087121

ABSTRACT

Rapid and precise serum cytokine quantification provides immense clinical significance in monitoring the immune status of patients in rapidly evolving infectious/inflammatory disorders, examplified by the ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. However, real-time information on predictive cytokine biomarkers to guide targetable immune pathways in pathogenic inflammation is critically lacking, because of the insufficient detection range and detection limit in current label-free cytokine immunoassays. In this work, we report a highly sensitive localized surface plasmon resonance imaging (LSPRi) immunoassay for label-free Interleukin 6 (IL-6) detection utilizing rationally designed peptide aptamers as the capture interface. Benefiting from its characteristically smaller dimension and direct functionalization on the sensing surface via Au-S bonding, the peptide-aptamer-based LSPRi immunoassay achieved enhanced label-free serum IL-6 detection with a record-breaking limit of detection down to 4.6 pg/mL, and a wide dynamic range of ∼6 orders of magnitude (values from 4.6 to 1 × 106 pg/mL were observed). The immunoassay was validated in vitro for label-free analysis of SARS-CoV-2 induced inflammation, and further applied in rapid quantification of serum IL-6 profiles in COVID-19 patients. Our peptide aptamer LSPRi immunoassay demonstrates great potency in label-free cytokine detection with unprecedented sensing capability to provide accurate and timely interpretation of the inflammatory status and disease progression, and determination of prognosis.


Subject(s)
Aptamers, Peptide , Biosensing Techniques , COVID-19 , Humans , SARS-CoV-2 , Cytokines/analysis , Interleukin-6 , Immunoassay/methods , Inflammation
8.
Biosensors (Basel) ; 12(10)2022 Sep 23.
Article in English | MEDLINE | ID: covidwho-2043580

ABSTRACT

The global pandemic of COVID-19 has created an unrivalled need for sensitive and rapid point-of-care testing (POCT) methods for the detection of infectious viruses. For the novel coronavirus SARS-CoV-2, the nucleocapsid protein (N-protein) is one of the most abundant structural proteins of the virus and it serves as a useful diagnostic marker for detection. Herein, we report a fiber optic particle plasmon resonance (FOPPR) biosensor which employed a single-stranded DNA (ssDNA) aptamer as the recognition element to detect the SARS-CoV-2 N-protein in 15 min with a limit of detection (LOD) of 2.8 nM, meeting the acceptable LOD of 106 copies/mL set by the WHO target product profile. The sensor chip is a microfluidic chip based on the balance between the gravitational potential and the capillary force to control fluid loading, thus enabling the power-free auto-flowing function. It also has a risk-free self-contained design to avoid the risk of the virus leaking into the environment. These findings demonstrate the potential for designing a low-cost and robust POCT device towards rapid antigen detection for early screening of SARS-CoV-2 and its related mutants.


Subject(s)
Biosensing Techniques , COVID-19 , Humans , SARS-CoV-2 , DNA, Single-Stranded , Microfluidics , COVID-19/diagnosis , Nucleocapsid Proteins/genetics
9.
Micromachines (Basel) ; 13(9)2022 Sep 14.
Article in English | MEDLINE | ID: covidwho-2033058

ABSTRACT

In recent years, the emergence of COVID-19 and other epidemics caused by RNA(ribonucleic acid)-type genetic viruses has aroused the close attention of governments around the world on emergency response to public safety and health emergencies. In this paper, an electrodeless biosensing detection chip for RNA virus medical detection is designed using quartz crystal microbalance technology and local surface plasmon resonance technology. The plasmonic resonance characteristic in the nanostructures of gold nanorods-quartz substrates with different parameters and the surface potential distribution of the quartz crystal microbalance sensing chip were studied by COMSOL finite element simulation software. The results show that the arrangement structure and spacing of gold nanorod dimers greatly affect the local surface plasmon resonance of nanorods, which in turn affects the detection results of biomolecules. Moreover, high concentrations of "hot spots" are distributed between both ends and the gap of the gold nanorod dimer, which reflects the strong hybridization of the multiple resonance modes of the nanoparticles. In addition, by simulating and calculating the surface potential distribution of the electrode area and non-electrode area of the biosensor chip, it was found that the biosensor chip with these two areas can enhance the piezoelectric effect of the quartz chip. Under the same simulation conditions, the biochip with a completely electrodeless structure showed a better sensing performance. The sensor chip combining QCM and LSPR can reduce the influence of the metal electrode on the quartz wafer to improve the sensitivity and accuracy of detection. Considering the significant influence of the gold nanorod dimer plasma resonance mode and the significant advantages of the electrodeless biosensor chip, an electrodeless biosensor combining these two technologies is proposed for RNA virus detection and screening, which has potential applications in biomolecular measurement and other related fields.

10.
Sens Actuators B Chem ; 373: 132638, 2022 Dec 15.
Article in English | MEDLINE | ID: covidwho-2031689

ABSTRACT

Stratifying patients according to disease severity has been a major hurdle during the COVID-19 pandemic. This usually requires evaluating the levels of several biomarkers, which may be cumbersome when rapid decisions are required. In this manuscript we show that a single nanoparticle aggregation test can be used to distinguish patients that require intensive care from those that have already been discharged from the intensive care unit (ICU). It consists of diluting a platelet-free plasma sample and then adding gold nanoparticles. The nanoparticles aggregate to a larger extent when the samples are obtained from a patient in the ICU. This changes the color of the colloidal suspension, which can be evaluated by measuring the pixel intensity of a photograph. Although the exact factor or combination of factors behind the different aggregation behavior is unknown, control experiments demonstrate that the presence of proteins in the samples is crucial for the test to work. Principal component analysis demonstrates that the test result is highly correlated to biomarkers of prognosis and inflammation that are commonly used to evaluate the severity of COVID-19 patients. The results shown here pave the way to develop nanoparticle aggregation assays that classify COVID-19 patients according to disease severity, which could be useful to de-escalate care safely and make a better use of hospital resources.

11.
Clinical and Translational Biophotonics, Translational 2022 ; 2022.
Article in English | Scopus | ID: covidwho-2012125

ABSTRACT

Localized surface plasmon resonance of Au nanodots array are very sensitive and resonance field disturbance due to 100 nm sized SARS-CoV-2 virus can be detected via resonance wavelength shift. We have proposed Au nanodots (100 nm diameter and 200 nm pitch) array plasmonic biosensing platform for SARS-CoV-2 virus detection. © 2022 The Author(s).

12.
Nanomaterials (Basel) ; 12(15)2022 Jul 29.
Article in English | MEDLINE | ID: covidwho-1994123

ABSTRACT

The work introduces a localized surface plasmon resonance (LSPR) sensor chip integrated with vertical-cavity surface-emitting lasers (VCSELs). Using VCSEL as the light source, the hexagonal gold nanoparticle array was integrated with anodic aluminum oxide (AAO) as the mask on the light-emitting end face. The sensitivity sensing test of the refractive index solution was realized, combined with microfluidic technology. At the same time, the finite-difference time- domain (FDTD) algorithm was applied to model and simulate the gold nanostructures. The experimental results showed that the output power of the sensor was related to the refractive index of the sucrose solution. The maximum sensitivity of the sensor was 1.65 × 106 nW/RIU, which gives it great application potential in the field of biomolecular detection.

13.
Chemical, Biological, Radiological, Nuclear, and Explosives (CBRNE) Sensing XXIII 2022 ; 12116, 2022.
Article in English | Scopus | ID: covidwho-1923081

ABSTRACT

A rapid, portable, and cost-effective method to detect the infection of SARS-CoV-2 is fundamental toward mitigating the current COVID-19 pandemic. A localized surface plasmon resonance (LSPR) sensor based on human angiotensin-converting enzyme 2 protein (ACE2) functionalized silver nanotriangle array is developed for rapid coronavirus detection. The sensor is validated by SARS-CoV-2 spike RBD protein and CoV NL63 virus with high sensitivity and specificity. A linear shift of the LSPR wavelength and transmission intensity at a fixed wavelength (750 nm) versus the logarithm of the concentration of the spike RBD protein and CoV NL63 is observed. The limits of detection for the spike RBD protein, CoV NL63 in untreated saliva are determined to be 0.38 pM, and 625 PFU/mL, respectively, while the detection time is found to be less than 20 min. Such a LSPR sensor could serve as a potential rapid point-of-care diagnostic platform for COVID-19. © 2022 SPIE

14.
Chemosensors ; 10(4):136, 2022.
Article in English | ProQuest Central | ID: covidwho-1809733

ABSTRACT

In the last few decades, plasmonic colorimetric biosensors raised increasing interest in bioanalytics thanks to their cost-effectiveness, responsiveness, and simplicity as compared to conventional laboratory techniques. Potential high-throughput screening and easy-to-use assay procedures make them also suitable for realizing point of care devices. Nevertheless, several challenges such as fabrication complexity, laborious biofunctionalization, and poor sensitivity compromise their technological transfer from research laboratories to industry and, hence, still hamper their adoption on large-scale. However, newly-developing plasmonic colorimetric biosensors boast impressive sensing performance in terms of sensitivity, dynamic range, limit of detection, reliability, and specificity thereby continuously encouraging further researches. In this review, recently reported plasmonic colorimetric biosensors are discussed with a focus on the following categories: (i) on-platform-based (localized surface plasmon resonance, coupled plasmon resonance and surface lattice resonance);(ii) colloid aggregation-based (label-based and label free);(iii) colloid non-aggregation-based (nanozyme, etching-based and growth-based).

15.
Biosensors (Basel) ; 12(3)2022 Feb 28.
Article in English | MEDLINE | ID: covidwho-1715109

ABSTRACT

Cost-effective, rapid, and sensitive detection of SARS-CoV-2, in high-throughput, is crucial in controlling the COVID-19 epidemic. In this study, we proposed a vertical microcavity and localized surface plasmon resonance hybrid biosensor for SARS-CoV-2 detection in artificial saliva and assessed its efficacy. The proposed biosensor monitors the valley shifts in the reflectance spectrum, as induced by changes in the refractive index within the proximity of the sensor surface. A low-cost and fast method was developed to form nanoporous gold (NPG) with different surface morphologies on the vertical microcavity wafer, followed by immobilization with the SARS-CoV-2 antibody for capturing the virus. Modeling and simulation were conducted to optimize the microcavity structure and the NPG parameters. Simulation results revealed that NPG-deposited sensors performed better in resonance quality and in sensitivity compared to gold-deposited and pure microcavity sensors. The experiment confirmed the effect of NPG surface morphology on the biosensor sensitivity as demonstrated by simulation. Pre-clinical validation revealed that 40% porosity led to the highest sensitivity for SARS-CoV-2 pseudovirus at 319 copies/mL in artificial saliva. The proposed automatic biosensing system delivered the results of 100 samples within 30 min, demonstrating its potential for on-site coronavirus detection with sufficient sensitivity.


Subject(s)
Biosensing Techniques , COVID-19 , COVID-19/diagnosis , Gold/chemistry , Humans , SARS-CoV-2 , Surface Plasmon Resonance
16.
Sens Actuators B Chem ; 359: 131604, 2022 May 15.
Article in English | MEDLINE | ID: covidwho-1692880

ABSTRACT

A rapid, portable, and cost-effective method to detect the infection of SARS-CoV-2 is fundamental toward mitigating the current COVID-19 pandemic. Herein, a human angiotensin-converting enzyme 2 protein (ACE2) functionalized silver nanotriangle (AgNT) array localized surface plasmon resonance (LSPR) sensor is developed for rapid coronavirus detection, which is validated by SARS-CoV-2 spike RBD protein and CoV NL63 virus with high sensitivity and specificity. A linear shift of the LSPR wavelength versus the logarithm of the concentration of the spike RBD protein and CoV NL63 is observed. The limits of detection for the spike RBD protein, CoV NL63 in buffer and untreated saliva are determined to be 0.83 pM, 391 PFU/mL, and 625 PFU/mL, respectively, while the detection time is found to be less than 20 min. Thus, the AgNT array optical sensor could serve as a potential rapid point-of-care COVID-19 diagnostic platform.

17.
Nanomaterials (Basel) ; 11(12)2021 Nov 24.
Article in English | MEDLINE | ID: covidwho-1590935

ABSTRACT

Monoclonal antibody-based immunotherapy is one of the pillars of cancer treatment. However, for an efficient and personalized approach to the therapy, a quantitative evaluation of the right dose for each patient is required. In this study, we developed a simple, label-free, and rapid approach to quantify Trastuzumab, a humanized IgG1 monoclonal antibody used against human epidermal growth factor receptor 2 (HER2), overexpressed in breast cancer patients, based on localized surface plasmon resonance (LSPR). The central idea of this work was to use gold nanoparticles (AuNPs) as plasmonic scaffolds, decorated with HER2 binders mixed with oligo-ethylene glycol (OEG) molecules, to tune the surface density of the attached macromolecules and to minimize nonspecific binding events. Specifically, we characterized and optimized a self-assembled monolayer of mixed alkylthiols terminated with nitrilotriacetic acid (NTA), and OEG3 as a spacing ligand to achieve both excellent dispersibility and high reliability in protein immobilization. The successful immobilization of histidine-tagged HER2 (His-tagged HER2) on NTA via cobalt (II) chelates was demonstrated, confirming the fully functional attachment of the proteins to the AuNP surface. The proposed design demonstrates the capability of producing a clear readout that enables the transduction of a Trastuzumab/HER2 binding event into optical signals based on the wavelength shifts in LSPR, which allowed for detecting clinically relevant concentrations of Trastuzumab down to 300 ng/mL in the buffer and 2 µg/mL in the diluted serum. This strategy was found to be fast and highly specific to Trastuzumab. These findings make the present platform an auspicious tool for developing affordable bio-nanosensors.

18.
Sensors and Actuators B: Chemical ; : 131327, 2021.
Article in English | ScienceDirect | ID: covidwho-1586490

ABSTRACT

Epstein-Barr virus (EBV) positivity is one of the indexes for diagnosing nasopharyngeal carcinoma (NPC). Moreover, systemic inflammatory responses can easily be triggered in patients who are both EBV- and coronavirus disease 2019 (COVID-19)-positive. Development of rapid and highly sensitive EBV screening methods has become important. In this study, a nanofluidic preconcentrator integrated with a nanoslit Fano resonance biosensor was developed to detect latent membrane protein 1 (LMP1) for an EBV diagnosis. Through nanoimprinting and aluminum deposition, the low-cost nanoslit plasmonic sensing chip can be mass-produced. The nanoporous membrane was patterned on a sensing chip as an ion selective channel to concentrate LMP1 proteins. Anti-LMP1 immunoglobulin G was then modified to a sensing chip to immunosense LMP1. The Fano resonant spectrum of the capped nanoslit array produced a transmission peak followed by a dip. We recorded and analyzed the spectrum using four methods, including area, center of mass, peak value, and dip value methods. With preconcentration, a limit of detection (LOD) of 100pg/ml and a sensing range of 100pg/ml to 10µg/ml was achieved using the peak value.

19.
Int J Mol Sci ; 22(19)2021 Oct 01.
Article in English | MEDLINE | ID: covidwho-1463707

ABSTRACT

The electron density of a nanoparticle is a very important characteristic of the properties of a material. This paper describes the formation of silver nanoparticles (NPs) and the variation in the electronic state of an NP's surface upon the reduction in Ag+ ions with oxalate ions, induced by UV irradiation. The calculations were based on optical spectrophotometry data. The NPs were characterized using Transmission electron microscopy and Dynamic light scattering. As ~10 nm nanoparticles are formed, the localized surface plasmon resonance (LSPR) band increases in intensity, decreases in width, and shifts to the UV region from 402 to 383 nm. The interband transitions (IBT) band (≤250 nm) increases in intensity, with the band shape and position remaining unchanged. The change in the shape and position of the LSPR band of silver nanoparticles in the course of their formation is attributable to an increasing concentration of free electrons in the particles as a result of a reduction in Ag+ ions on the surface and electron injection by CO2- radicals. The ζ-potential of colloids increases with an increase in electron density in silver nuclei. A quantitative relationship between this shift and electron density on the surface was derived on the basis of the Mie-Drude theory. The observed blue shift (19 nm) corresponds to an approximately 10% increase in the concentration of electrons in silver nanoparticles.


Subject(s)
Electricity , Electrons , Metal Nanoparticles/chemistry , Silver/chemistry , Solutions/chemistry , Chemical Phenomena , Electrochemistry , Metal Nanoparticles/ultrastructure , Microscopy, Electron, Transmission , Models, Theoretical , Particle Size , Surface Plasmon Resonance
20.
Microchem J ; 167: 106305, 2021 Aug.
Article in English | MEDLINE | ID: covidwho-1198979

ABSTRACT

Since December 2019, we have been in the battlefield with a new threat to the humanity known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this review, we describe the four main methods used for diagnosis, screening and/or surveillance of SARS-CoV-2: Real-time reverse transcription polymerase chain reaction (RT-PCR); chest computed tomography (CT); and different complementary alternatives developed in order to obtain rapid results, antigen and antibody detection. All of them compare the highlighting advantages and disadvantages from an analytical point of view. The gold standard method in terms of sensitivity and specificity is the RT-PCR. The different modifications propose to make it more rapid and applicable at point of care (POC) are also presented and discussed. CT images are limited to central hospitals. However, being combined with RT-PCR is the most robust and accurate way to confirm COVID-19 infection. Antibody tests, although unable to provide reliable results on the status of the infection, are suitable for carrying out maximum screening of the population in order to know the immune capacity. More recently, antigen tests, less sensitive than RT-PCR, have been authorized to determine in a quicker way whether the patient is infected at the time of analysis and without the need of specific instruments.

SELECTION OF CITATIONS
SEARCH DETAIL