Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Assunto da revista
Intervalo de ano de publicação
1.
J Pediatr ; 257: 113358, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36822507

RESUMO

Using an electronic health record-based algorithm, we identified children with Coronavirus disease 2019 (COVID-19) based exclusively on serologic testing between March 2020 and April 2022. Compared with the 131 537 polymerase chain reaction-positive children, the 2714 serology-positive children were more likely to be inpatients (24% vs 2%), to have a chronic condition (37% vs 24%), and to have a diagnosis of multisystem inflammatory syndrome in children (23% vs <1%). Identification of children who could have been asymptomatic or paucisymptomatic and not tested is critical to define the burden of post-acute sequelae of severe acute respiratory syndrome coronavirus 2 infection in children.


Assuntos
COVID-19 , Humanos , Criança , COVID-19/complicações , COVID-19/diagnóstico , Síndrome de COVID-19 Pós-Aguda , SARS-CoV-2 , Estudos de Coortes , Registros Eletrônicos de Saúde , Anticorpos Antivirais , Progressão da Doença , Teste para COVID-19
2.
Anal Bioanal Chem ; 415(3): 391-404, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36346456

RESUMO

The SARS-CoV-2 pandemic has shown the importance of rapid and comprehensive diagnostic tools. While there are numerous rapid antigen tests available, rapid serological assays for the detection of neutralizing antibodies are and will be needed to determine not only the amount of antibodies formed after infection or vaccination but also their neutralizing potential, preventing the cell entry of SARS-CoV-2. Current active-virus neutralization assays require biosafety level 3 facilities, while virus-free surrogate assays are more versatile in applications, but still take typically several hours until results are available. To overcome these disadvantages, we developed a competitive chemiluminescence immunoassay that enables the detection of neutralizing SARS-CoV-2 antibodies within 7 min. The neutralizing antibodies bind to the viral receptor binding domain (RBD) and inhibit the binding to the human angiotensin-converting enzyme 2 (ACE2) receptor. This competitive binding inhibition test was characterized with a set of 80 samples, which could all be classified correctly. The assay results favorably compare to those obtained with a more time-intensive ELISA-based neutralization test and a commercial surrogate neutralization assay. Our test could further be used to detect individuals with a high total IgG antibody titer, but only a low neutralizing titer, as well as for monitoring neutralizing antibodies after vaccinations. This effective performance in SARS-CoV-2 seromonitoring delineates the potential for the test to be adapted to other diseases in the future.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/diagnóstico , Ensaio de Imunoadsorção Enzimática , Imunoensaio , Luminescência , Automação Laboratorial
3.
Anal Bioanal Chem ; 413(22): 5619-5632, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33983466

RESUMO

In the face of the COVID-19 pandemic, the need for rapid serological tests that allow multiplexing emerged, as antibody seropositivity can instruct about individual immunity after an infection with SARS-CoV-2 or after vaccination. As many commercial antibody tests are either time-consuming or tend to produce false negative or false positive results when only one antigen is considered, we developed an automated, flow-based chemiluminescence microarray immunoassay (CL-MIA) that allows for the detection of IgG antibodies to SARS-CoV-2 receptor-binding domain (RBD), spike protein (S1 fragment), and nucleocapsid protein (N) in human serum and plasma in less than 8 min. The CoVRapid CL-MIA was tested with a set of 65 SARS-CoV-2 serology positive or negative samples, resulting in 100% diagnostic specificity and 100% diagnostic sensitivity, thus even outcompeting commercial tests run on the same sample set. Additionally, the prospect of future quantitative assessments (i.e., quantifying the level of antibodies) was demonstrated. Due to the fully automated process, the test can easily be operated in hospitals, medical practices, or vaccination centers, offering a valuable tool for COVID-19 serosurveillance. Graphical abstract.


Assuntos
Anticorpos Antivirais/sangue , Teste Sorológico para COVID-19/métodos , Imunoensaio/métodos , Imunoglobulina G/sangue , SARS-CoV-2/imunologia , Antígenos Virais/química , Antígenos Virais/imunologia , Automação Laboratorial , Proteínas do Nucleocapsídeo de Coronavírus/imunologia , Humanos , Proteínas Imobilizadas/química , Proteínas Imobilizadas/imunologia , Soros Imunes , Imunoensaio/instrumentação , Dispositivos Lab-On-A-Chip , Medições Luminescentes , Fosfoproteínas/imunologia , Sensibilidade e Especificidade , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Fatores de Tempo
4.
Eur J Clin Invest ; 50(10): e13357, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32691863

RESUMO

AIMS: To validate the diagnostic accuracy of the Augurix SARS-CoV-2 IgM/IgG rapid immunoassay diagnostic test (RDT) for COVID-19. METHODS: In this unmatched 1:1 case-control study, blood samples from 46 real-time RT-PCR-confirmed SARS-CoV-2 hospitalized cases and 45 healthy donors (negative controls) were studied. Diagnostic accuracy of the IgG RDT was assessed against both an in-house recombinant spike-expressing immunofluorescence assay (rIFA), as an established reference method (primary endpoint), and the Euroimmun SARS-CoV-2 IgG enzyme-linked immunosorbent assays (ELISA) (secondary endpoint). RESULTS: COVID-19 patients were more likely to be male (61% vs 20%; P = .0001) and older (median 66 vs 47 years old; P < .001) than controls. Whole blood IgG-RDT results showed 86% and 93% overall Kendall concordance with rIFA and IgG ELISA, respectively. IgG RDT performances were similar between plasma and whole blood. Overall, RDT sensitivity was 88% (95% confidence interval [95%CI]: 70-96), specificity 98% (95%CI: 90-100), PPV 97% (95%CI: 80-100) and NPV 94% (95%CI: 84-98). The IgG-RDT carried out from 0 to 6 days, 7 to 14 days and > 14 days after the SARS-CoV-2 RT-PCR test displayed 30%, 73% and 100% positivity rates in the COVID-19 group, respectively. When considering samples taken >14 days after RT-PCR diagnosis, NPV was 100% (95%CI:90-100), and PPV was 100% (95%CI:72-100). CONCLUSIONS: The Augurix IgG-RDT done in whole blood displays a high diagnostic accuracy for SARS-CoV-2 IgG in high COVID-19 prevalence settings, where its use could be considered in the absence of routine diagnostic serology facilities.


Assuntos
Anticorpos Antivirais/imunologia , Betacoronavirus/imunologia , Infecções por Coronavirus/diagnóstico , Pneumonia Viral/diagnóstico , Testes Sorológicos , Glicoproteína da Espícula de Coronavírus/imunologia , Idoso , COVID-19 , Teste para COVID-19 , Vacinas contra COVID-19 , Estudos de Casos e Controles , Técnicas de Laboratório Clínico , Feminino , Imunofluorescência , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Masculino , Pessoa de Meia-Idade , Pandemias , Reação em Cadeia da Polimerase Via Transcriptase Reversa , SARS-CoV-2 , Sensibilidade e Especificidade
5.
Talanta ; 271: 125685, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38262129

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic recently demonstrated the devastating impact on public health, economy, and social development of zoonotic infectious diseases, whereby viruses jump from animals to infect humans. Due to this potential of viruses to cross the species barrier, the surveillance of infectious pathogens circulation in domestic and close-to-human animals is indispensable, as they could be potential reservoirs. Optical biosensors, mainly those based on Surface Plasmon Resonance (SPR), have widely demonstrated its ability for providing direct, label-free, and quantitative bioanalysis with excellent sensitivity and reliability. This biosensor technology can provide a powerful tool to the veterinary field, potentially being helpful for the monitoring of the infection spread. We have implemented a multi-target COVID-19 serology plasmonic biosensor for the rapid testing and screening of common European domestic animals. The multi-target serological biosensor assay enables the detection of total SARS-CoV-2 antibodies (IgG + IgM) generated towards both S and N viral antigens. The analysis is performed in less than 15 min with a low-volume serum sample (<20 µL, 1:10 dilution), reaching a limit of detection of 49.6 ng mL-1. A complete validation has been carried out with hamster, dog, and cat sera samples (N = 75, including 37 COVID-19-positive and 38 negative samples). The biosensor exhibits an excellent diagnostic sensitivity (100 %) and good specificity (71.4 %) for future application in veterinary settings. Furthermore, the biosensor technology is integrated into a compact, portable, and user-friendly device, well-suited for point-of-care testing. This study positions our plasmonic biosensor as an alternative and reliable diagnostic tool for COVID-19 serology in animal samples, expanding the applicability of plasmonic technologies for decentralized analysis in veterinary healthcare and animal research.


Assuntos
Técnicas Biossensoriais , COVID-19 , Humanos , Animais , Cães , Gatos , COVID-19/diagnóstico , COVID-19/veterinária , SARS-CoV-2 , Animais Domésticos , Reprodutibilidade dos Testes , Anticorpos Antivirais , Sensibilidade e Especificidade , Teste para COVID-19
6.
Biotechnol Rep (Amst) ; 37: e00780, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36619904

RESUMO

SARS-CoV-2 receptor binding domain (RBD) recognizes the angiotensin converting enzyme 2 (ACE2) receptor in host cells that enables infection. Due to its antigenic specificity, RBD production is important for development of serological assays. Here we have established a system for stable RBD production in HEK 293T mammalian cells that simultaneously express the recombinant fluorescent protein dTomato, which enables kinetic monitoring of RBD expression by fluorescence microscopy. In addition, we have validated the use of this recombinant RBD in an ELISA assay for the detection of anti-RBD antibodies in serum samples of COVID-19 convalescent patients. Recombinant RBD generated using this approach can be useful for generation of antibody-based therapeutics against SARS-CoV-2, as well serological assays aimed to test antibody responses to this relevant virus.

7.
Vaccines (Basel) ; 11(3)2023 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-36992182

RESUMO

(1) Background: The high incidence of SARS-CoV-2 infection in vaccinated persons underscores the importance of individualized re-vaccination. PanIg antibodies that act against the S1/-receptor binding domain quantified in serum by a routine diagnostic test (ECLIA, Roche) can be used to gauge the individual ex vivo capacity of SARS-CoV-2 neutralization. However, that test is not adapted to mutations in the S1/-receptor binding domain, having accumulated in SARS-CoV-2 variants. Therefore, it might be unsuited to determine immune-reactivity against SARS-CoV-2 BA.5.1. (2) Method: To address this concern, we re-investigated sera obtained six months after second vaccinations with un-adapted mRNA vaccine Spikevax (Moderna). We related serum levels of panIg against the S1/-receptor binding domain quantified by the un-adapted ECLIA with full virus neutralization capacity against SARS-CoV-2 B.1 or SARS-CoV-2 BA5.1. (3) Results: 92% of the sera exhibited sufficient neutralization capacity against the B.1 strain. Only 20% of the sera sufficiently inhibited the BA5.1 strain. Sera inhibiting BA5.1 could not be distinguished from non-inhibiting sera by serum levels of panIg against the S1/-receptor binding domain quantified by the un-adapted ECLIA. (4) Conclusion: Quantitative serological tests for an antibody against the S1/-receptor binding domain are unsuited as vaccination companion diagnostics, unless they are regularly adapted to mutations that have accumulated in that domain.

8.
Clin Biochem ; 104: 66-69, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35131260

RESUMO

Salivary matrix is an appealing specimen type for SARS-CoV-2 serology because of ease of collection and potential for concurrent nucleic acid testing. We address the feasibility of salivary matrix to detect anti-SARS-CoV-2 antibodies using two commercially available anti-SARS-CoV-2 Total antibody assays including analytical validations. Matched serum and saliva samples were collected from 10 convalescent COVID-19 patients and tested using a quantitative anti-Spike Total antibody assay and a qualitative anti-Nucleocapsid Total antibody assay from Roche Diagnostics. Both assays were 100% sensitive for COVID-19 history in serum. However, saliva samples were below serum positivity thresholds. We then collected longitudinal salivary samples from a volunteer cohort receiving the Pfizer-BioNTech COVID-19 BNT162b2 vaccine. Saliva was negative for anti-SARS-CoV-2 antibodies at 5 time points after a single dose of vaccine including day 56 when mean (min-max) serum levels of anti-Spike Total antibody were 79.0 U/mL (46.6-110.1) (N = 8). After a second vaccine dose serum-matched samples were beyond the analytical measuring range of the assay (>2500 U/mL), and detection of salivary anti-Spike Total antibody was achieved in all volunteers (12.2 U/mL [2.0-32.7]) (N = 11) 30 days after the second dose. Mean anti-Spike Total antibody levels in serum (1558 U/mL (434->2500)) and saliva (2.6 U/mL (<0.4-11.4)) declined 216-233 days after the first dose of vaccine (P < 0.05); and saliva was 75% sensitive for two doses of vaccination at this latter time point (N = 25). These data suggest commercial assays are capable of detecting vaccine status after two doses of BNT162b2 vaccine up to 6 months and could inform COVID-19 surveillance.


Assuntos
COVID-19 , SARS-CoV-2 , Anticorpos Antivirais , Vacina BNT162 , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Humanos
9.
Int Health ; 14(1): 18-52, 2022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33620427

RESUMO

As the coronavirus disease 2019 (COVID-19) pandemic continues to rise and second waves are reported in some countries, serological test kits and strips are being considered to scale up an adequate laboratory response. This study provides an update on the kinetics of humoral immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and performance characteristics of serological protocols (lateral flow assay [LFA], chemiluminescence immunoassay [CLIA] and ELISA) used for evaluations of recent and past SARS-CoV-2 infection. A thorough and comprehensive review of suitable and eligible full-text articles was performed on PubMed, Scopus, Web of Science, Wordometer and medRxiv from 10 January to 16 July 2020. These articles were searched using the Medical Subject Headings terms 'COVID-19', 'Serological assay', 'Laboratory Diagnosis', 'Performance characteristics', 'POCT', 'LFA', 'CLIA', 'ELISA' and 'SARS-CoV-2'. Data from original research articles on SARS-CoV-2 antibody detection ≥second day postinfection were included in this study. In total, there were 7938 published articles on humoral immune response and laboratory diagnosis of COVID-19. Of these, 74 were included in this study. The detection, peak and decline period of blood anti-SARS-CoV-2 IgM, IgG and total antibodies for point-of-care testing (POCT), ELISA and CLIA vary widely. The most promising of these assays for POCT detected anti-SARS-CoV-2 at day 3 postinfection and peaked on the 15th day; ELISA products detected anti-SARS-CoV-2 IgM and IgG at days 2 and 6 then peaked on the eighth day; and the most promising CLIA product detected anti-SARS-CoV-2 at day 1 and peaked on the 30th day. The most promising LFA, ELISA and CLIA that had the best performance characteristics were those targeting total SARS-CoV-2 antibodies followed by those targeting anti-SARS-CoV-2 IgG then IgM. Essentially, the CLIA-based SARS-CoV-2 tests had the best performance characteristics, followed by ELISA then POCT. Given the varied performance characteristics of all the serological assays, there is a need to continuously improve their detection thresholds, as well as to monitor and re-evaluate their performances to assure their significance and applicability for COVID-19 clinical and epidemiological purposes.


Assuntos
COVID-19 , Humanos , Cinética , Pandemias , SARS-CoV-2 , Sensibilidade e Especificidade
10.
Biosens Bioelectron ; 213: 114476, 2022 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-35716642

RESUMO

Coronavirus disease 2019 (COVID-19) has caused significant global morbidity and mortality. The serology test that detects antibodies against the disease causative agent, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has often neglected value in supporting immunization policies and therapeutic decision-making. The ELISA-based antibody test is time-consuming and bulky. This work described a gold micro-interdigitated electrodes (IDE) biosensor for COVID antibody detection based on Electrochemical Impedance Spectroscopy (EIS) responses. The IDE architecture allows easy surface modification with the viral structure protein, Spike (S) protein, in the gap of the electrode digits to selectively capture anti-S antibodies in buffer solutions or human sera. Two strategies were employed to resolve the low sensitivity issue of non-faradic impedimetric sensors and the sensor fouling phenomenon when using the serum. One uses secondary antibody-gold nanoparticle (AuNP) conjugates to further distinguish anti-S antibodies from the non-specific binding and obtain a more significant impedance change. The second strategy consists of increasing the concentration of target antibodies in the gap of IDEs by inducing an AC electrokinetic effect such as dielectrophoresis (DEP). AuNP and DEP methods reached a limit of detection of 200 ng/mL and 2 µg/mL, respectively using purified antibodies in buffer, while the DEP method achieved a faster testing time of only 30 min. Both strategies could qualitatively distinguish COVID-19 antibody-positive and -negative sera. Our work, especially the impedimetric detection of COVID-19 antibodies under the assistance of the DEP force presents a promising path toward rapid, point-of-care solutions for COVID-19 serology tests.


Assuntos
Técnicas Biossensoriais , COVID-19 , Nanopartículas Metálicas , Técnicas Biossensoriais/métodos , COVID-19/diagnóstico , Eletrodos , Ouro/química , Humanos , Nanopartículas Metálicas/química , SARS-CoV-2
11.
Vaccines (Basel) ; 10(7)2022 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-35891208

RESUMO

Purpose: We describe a diagnostic procedure suitable for scheduling (re-)vaccination against severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) according to individual state of humoral immunization. Methods: To clarify the relation between quantitative antibody measurements and humoral ex vivo immune responsiveness, we monitored 124 individuals before, during and six months after vaccination with Spikevax (Moderna, Cambridge, MA, USA). Antibodies against SARS-CoV-2 spike (S1) protein receptor-binding domain (S1-AB) and against nucleocapsid antigens were measured by chemiluminescent immunoassay (Roche). Virus-neutralizing activities were determined by surrogate assays (NeutraLISA, Euroimmune; cPass, GenScript). Neutralization of SARS-CoV-2 in cell culture (full virus NT) served as an ex vivo correlate for humoral immune responsiveness. Results: Vaccination responses varied considerably. Six months after the second vaccination, participants still positive for the full virus NT were safely determined by S1-AB levels ≥1000 U/mL. The full virus NT-positive fraction of participants with S1-AB levels <1000 U/mL was identified by virus-neutralizing activities >70% as determined by surrogate assays (NeutraLISA or cPas). Participants that were full virus NT-negative and presumably insufficiently protected could thus be identified by a sensitivity of >83% and a specificity of >95%. Conclusion: The described diagnostic strategy possibly supports individualized (re-)vaccination schedules based on simple and rapid measurement of serum-based SARS-CoV-2 antibody levels. Our data apply only to WUHAN-type SARS-CoV-2 virus and the current version of the mRNA vaccine from Moderna (Cambridge, MA, USA). Adaptation to other vaccines and more recent SARS-CoV-2 strains will require modification of cut-offs and re-evaluation of sensitivity/specificity.

12.
Pathology ; 53(6): 773-779, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34412859

RESUMO

Serological assays for SARS-CoV-2 infection are now widely available for use in diagnostic laboratories. Limited data are available on the performance characteristics in different settings, and at time periods remote from the initial infection. Validation of the Abbott (Architect SARS-CoV-2 IgG), DiaSorin (Liaison SARS-CoV-2 S1/S2 IgG) and Roche (Cobas Elecsys Anti-SARS-CoV-2) assays was undertaken utilising 217 serum samples from 131 participants up to 7 months following COVID-19 infection. The Abbott and DiaSorin assays were implemented into routine laboratory workflow, with outcomes reported for 2764 clinical specimens. Sensitivity and specificity were concordant with the range reported by the manufacturers for all assays. Sensitivity across the convalescent period was highest for the Roche at 95.2-100% (95% CI 81.0-100%), then the DiaSorin at 88.1-100% (95% CI 76.0-100%), followed by the Abbott 68.2-100% (95% CI 53.4-100%). Sensitivity of the Abbott assay fell from approximately 5 months; on this assay paired serum samples for 45 participants showed a significant drop in the signal-to-cut-off ratio and 10 sero-reversion events. When used in clinical practice, all samples testing positive by both DiaSorin and Abbott assays were confirmed as true positive results. In this low prevalence setting, despite high laboratory specificity, the positive predictive value of a single positive assay was low. Comprehensive validation of serological assays is necessary to determine the optimal assay for each diagnostic setting. In this low prevalence setting we found implementation of two assays with different antibody targets maximised sensitivity and specificity, with confirmatory testing necessary for any sample which was positive in only one assay.


Assuntos
Anticorpos Antivirais/análise , Teste Sorológico para COVID-19/métodos , COVID-19/diagnóstico , Anticorpos Antivirais/sangue , Humanos , Laboratórios , Estudos Longitudinais , SARS-CoV-2 , Sensibilidade e Especificidade
13.
Healthcare (Basel) ; 9(9)2021 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-34574897

RESUMO

BACKGROUND: COVID-19 antibody testing has been shown to be predictive of prior COVID-19 infection and an effective testing tool. The CLUNGENE® SARS-COV-2 VIRUS (COVID-19) IgG/IgM Rapid Test Cassette was evaluated for its utility to aide healthcare professionals. METHOD: Two studies were performed by using the CLUNGENE® Rapid Test. (1) An expanded Point-of-Care (POC) study at two clinical sites was conducted to evaluate 99 clinical subjects: 62 positive subjects and 37 negative subjects were compared to RT-PCR, PPA, and NPA (95% CI). Sensitivity was calculated from blood-collection time following symptom onset. (2) A cross-reactivity study was performed to determine the potential for false-positive results from other common infections. RESULTS: The specificity of subjects with confirmed negative COVID-19 by RT-PCR was 100% (95% CI, 88.4-100.0%). The sensitivity of subjects with confirmed positive COVID-19 by RT-PCR was 96.77% (95% CI, 88.98-99.11%). In the cross-reactivity study, there were no false-positive results due to past infections or vaccinations unrelated to the SARS-CoV-2 virus. CONCLUSION: There is a need for a rapid, user-friendly, and inexpensive on-site monitoring system for diagnosis. The CLUNGENE® Rapid Test is a useful diagnostic test that provides results within 15 min, without high-complexity laboratory instrumentation.

14.
Clin Lab Med ; 40(4): 603-614, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33121625

RESUMO

The entire spectrum of diagnostic testing, from reagent supply to test performance, has been a major focus during the coronavirus disease 2019 (COVID-19) pandemic. The hope for serologic testing is that it will provide both epidemiologic information about seroprevalence as well as individual information about previous infection. This information is particularly helpful for high-risk individuals who may be outside of the viral shedding window, such as children with suspected multisystem inflammatory syndrome. It is not yet understood whether serologic testing can be interpreted in terms of protective immunity. These concerns must be addressed using highly sensitive and specific tests.


Assuntos
Betacoronavirus , Técnicas de Laboratório Clínico/métodos , Infecções por Coronavirus , Pandemias , Pneumonia Viral , Estudos Soroepidemiológicos , Betacoronavirus/imunologia , Betacoronavirus/isolamento & purificação , COVID-19 , Teste para COVID-19 , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Humanos , Imunidade Humoral/imunologia , Pandemias/prevenção & controle , Pneumonia Viral/epidemiologia , Pneumonia Viral/imunologia , Pneumonia Viral/prevenção & controle , Fatores de Proteção , SARS-CoV-2 , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA