Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nat Commun ; 14(1): 1772, 2023 03 30.
Article in English | MEDLINE | ID: covidwho-2279717

ABSTRACT

Several millions of individuals are estimated to develop post-acute sequelae SARS-CoV-2 condition (PASC) that persists for months after infection. Here we evaluate the immune response in convalescent individuals with PASC compared to convalescent asymptomatic and uninfected participants, six months following their COVID-19 diagnosis. Both convalescent asymptomatic and PASC cases are characterised by higher CD8+ T cell percentages, however, the proportion of blood CD8+ T cells expressing the mucosal homing receptor ß7 is low in PASC patients. CD8 T cells show increased expression of PD-1, perforin and granzyme B in PASC, and the plasma levels of type I and type III (mucosal) interferons are elevated. The humoral response is characterized by higher levels of IgA against the N and S viral proteins, particularly in those individuals who had severe acute disease. Our results also show that consistently elevated levels of IL-6, IL-8/CXCL8 and IP-10/CXCL10 during acute disease increase the risk to develop PASC. In summary, our study indicates that PASC is defined by persisting immunological dysfunction as late as six months following SARS-CoV-2 infection, including alterations in mucosal immune parameters, redistribution of mucosal CD8+ß7Integrin+ T cells and IgA, indicative of potential viral persistence and mucosal involvement in the etiopathology of PASC.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Acute Disease , CD8-Positive T-Lymphocytes , COVID-19 Testing , Disease Progression , Immunoglobulin A
2.
Acta Med Port ; 36(7-8): 506-510, 2023 Jul 03.
Article in English | MEDLINE | ID: covidwho-2217832

ABSTRACT

Several cases of paediatric acute hepatitis of an unknown aetiology have been described in these last few months and in several countries worldwide. We present two patients, a 7-month-old girl and an 8-year-old boy, with gastrointestinal symptoms and lethargy, associated with elevation of transaminase levels. Serologies for hepatitis A-E virus and PCR test to SARS-CoV-2 were all negative. In the first case, an adenovirus serotype C could be isolated in a respiratory sample as well as cytomegalovirus (CMV) in the blood (100 copies/mL). In both children, there was a progressive decrease in the hepatic markers and symptomatic resolution, compatible with a good prognosis, also seen globally in most cases. To date, infection remains the most plausible cause to consider, especially when it is presumed to be linked to adenovirus. Other potential agents and causes are still being evaluated, thus emphasizing the importance of continuous epidemiological surveillance, notification, and detailed study of all hepatitis cases.


Subject(s)
COVID-19 , Hepatitis A virus , Hepatitis A , Male , Female , Child , Humans , Infant , Portugal/epidemiology , SARS-CoV-2 , Hepatitis A/epidemiology , Acute Disease
3.
Lancet Respir Med ; 11(2): 163-175, 2023 02.
Article in English | MEDLINE | ID: covidwho-2184778

ABSTRACT

BACKGROUND: To inform future research and practice, we aimed to investigate the outcomes of patients who received extracorporeal membrane oxygenation (ECMO) for acute respiratory distress syndrome (ARDS) due to different variants of SARS-CoV-2. METHODS: This retrospective study included consecutive adult patients with laboratory-confirmed SARS-CoV-2 infection who received ECMO for ARDS in 21 experienced ECMO centres in eight European countries (Austria, Belgium, England, France, Germany, Italy, Portugal, and Spain) between Jan 1, 2020, and Sept 30, 2021. We collected data on patient characteristics, clinical status, and management before and after the initiation of ECMO. Participants were grouped according to SARS-CoV-2 variant (wild type, alpha, delta, or other) and period of the pandemic (first [Jan 1-June 30] and second [July 1-Dec 31] semesters of 2020, and first [Jan 1-June 30] and second [July 1-Sept 30] semesters of 2021). Descriptive statistics and Kaplan-Meier survival curves were used to analyse evolving characteristics, management, and patient outcomes over the first 2 years of the pandemic, and independent risk factors of mortality were determined using multivariable Cox regression models. The primary outcome was mortality 90 days after the initiation of ECMO, with follow-up to Dec 30, 2021. FINDINGS: ECMO was initiated in 1345 patients. Patient characteristics and management were similar for the groups of patients infected with different variants, except that those with the delta variant had a younger median age and less hypertension and diabetes. 90-day mortality was 42% (569 of 1345 patients died) overall, and 43% (297/686) in patients infected with wild-type SARS-CoV-2, 39% (152/391) in those with the alpha variant, 40% (78/195) in those with the delta variant, and 58% (42/73) in patients infected with other variants (mainly beta and gamma). Mortality was 10% higher (50%) in the second semester of 2020, when the wild-type variant was still prevailing, than in other semesters (40%). Independent predictors of mortality were age, immunocompromised status, a longer time from intensive care unit admission to intubation, need for renal replacement therapy, and higher Sequential Organ Failure Assessment haemodynamic component score, partial pressure of arterial carbon dioxide, and lactate concentration before ECMO. After adjusting for these variables, mortality was significantly higher with the delta variant than with the other variants, the wild-type strain being the reference. INTERPRETATION: Although crude mortality did not differ between variants, adjusted risk of death was highest for patients treated with ECMO infected with the delta variant of SARS-CoV-2. The higher virulence and poorer outcomes associated with the delta strain might relate to higher viral load and increased inflammatory response syndrome in infected patients, reinforcing the need for a higher rate of vaccination in the population and updated selection criteria for ECMO, should a new and highly virulent strain of SARS-CoV-2 emerge in the future. Mortality was noticeably lower than in other large, multicentre series of patients who received ECMO for COVID-19, highlighting the need to concentrate resources at experienced centres. FUNDING: None.


Subject(s)
COVID-19 , Extracorporeal Membrane Oxygenation , Respiratory Distress Syndrome , Adult , Humans , SARS-CoV-2 , COVID-19/epidemiology , COVID-19/therapy , COVID-19/etiology , Retrospective Studies , Extracorporeal Membrane Oxygenation/adverse effects , Pandemics
4.
Cureus ; 14(10): e30599, 2022 Oct.
Article in English | MEDLINE | ID: covidwho-2124086

ABSTRACT

INTRODUCTION: In coronavirus disease 2019 (COVID-19), there are no tools available for the difficult task of recognizing which patients do not benefit from maintaining respiratory support, such as noninvasive ventilation (NIV). Identifying treatment failure is crucial to provide the best possible care and optimizing resources. Therefore, this study aimed to build a model that predicts NIV failure in patients who did not progress to invasive mechanical ventilation (IMV). METHODS: This retrospective observational study included critical COVID-19 patients treated with NIV who did not progress to IMV. Patients were admitted to a Portuguese tertiary hospital between October 1, 2020, and March 31, 2021. The outcome of interest was NIV failure, defined as COVID-19-related in-hospital death. A binary logistic regression was performed, where the outcome (mortality) was the dependent variable. Using the independent variables of the logistic regression a decision-tree classification model was implemented. RESULTS: The study sample, composed of 103 patients, had a mean age of 66.3 years (SD=14.9), of which 38.8% (40 patients) were female. Most patients (82.5%) were autonomous for basic activities of daily living. The prediction model was statistically significant with an area under the curve of 0.994 and a precision of 0.950. Higher age, a higher number of days with increases in the fraction of inspired oxygen (FiO2), a higher number of days of maximum expiratory positive airway pressure, a lower number of days on NIV, and a lower number of days from disease onset to hospital admission were, with statistical significance, associated with increased odds of death. A decision-tree classification model was then obtained to achieve the best combination of variables to predict the outcome of interest. CONCLUSIONS: This study presents a model to predict death in COVID-19 patients treated with NIV in patients who did not progress to IMV, based on easily applicable variables that mainly reflect patients' evolution during hospitalization. Along with the decision-tree classification model, these original findings may help clinicians define the best therapeutical approach to each patient, prioritizing life-comforting measures when adequate, and optimizing resources, which is crucial within limited or overloaded healthcare systems. Further research is needed on this subject of treatment failure, not only to understand if these results are reproducible but also, in a broader sense, helping to fill this gap in modern medicine guidelines.

5.
Vaccines (Basel) ; 10(11)2022 Nov 15.
Article in English | MEDLINE | ID: covidwho-2116046

ABSTRACT

We conducted a prospective study to evaluate immune responses to SARS-CoV-2 in oncology workers in which we collected blood and clinical data every 6 months. Spike-specific CD4+ T-cells and immunoglobulin G responses were measured using interferon-gamma enzyme-linked immunosorbent spot and enzyme-linked immunosorbent assay, respectively. Sixty (81%) vaccinated and 14 (19%) unvaccinated individuals were enrolled. CD4+ T-cell responses of those individuals currently naturally infected were comparable to those who were 6 months from receiving their last dose of the vaccine; both responses were significantly higher than among those who were unvaccinated. Unvaccinated participants who became vaccinated while in the study showed a significant increase in both types of spike-specific immune responses. Previously vaccinated individuals who received a third dose (booster) showed a similar response to the spike protein. However, this response decreases as soon as 3 months but does not dip below the established response following two doses. Response to variants of concern B.1.617.2 (Delta) and B.1.1.529 (Omicron) also increased, with the Omicron variant having a significantly lower response when compared to Delta and the wild type. We conclude that antibody and T-cell responses increase in oncology workers after serial vaccination but can wane over time.

6.
Med Clin (Engl Ed) ; 158(12): 569-575, 2022 Jun 24.
Article in English | MEDLINE | ID: covidwho-2117684

ABSTRACT

Introduction and purpose: Atrial fibrillation (AF) is common in patients admitted with severe COVID-19. However, there is limited data about the management of chronic anticoagulation therapy in these patients. We assessed the anticoagulation and incidence of major cardiovascular events in hospitalized patients with AF and COVID-19. Methods: We retrospectively investigated all consecutive patients with AF admitted with COVID-19 between March and May 2020 in 9 Spanish hospitals. We selected a control group of non-AF patients consecutively admitted with COVID-19. We compared baseline characteristics, incidence of major bleeding, thrombotic events and mortality. We used propensity score matching (PSM) to minimize potential confounding variables, as well as a multivariate analysis to predict major bleeding and death. Results: 305 patients admitted with AF and COVID-19 were included. After PSM, 151 AF patients were matched with 151 control group patients. During admission, low-molecular-weight heparin was the principal anticoagulant and the incidence of major bleeding and mortality were higher in the AF group [16 (10.6%) vs 3 (2%), p = 0.003; 52 (34.4%) vs 35 (23.2%), p = 0.03, respectively]. The multivariate analysis showed the presence of AF as independent predictor of in-hospital major bleeding and mortality in COVID-19 patients. In AF group, a secondary multivariate analysis identified high levels of D-dimer as independent predictor of in-hospital major bleeding. Conclusions: AF patients admitted with COVID-19 represent a population at high risk for bleeding and mortality during admission. It seems advisable to individualize anticoagulation therapy during admission, considering patient specific bleeding and thrombotic risk.


Antecedentes y objetivos: La fibrilación auricular (FA) es frecuente en pacientes ingresados por COVID-19 grave. Sin embargo, los datos sobre el manejo de la anticoagulación crónica en estos pacientes son escasos. Analizamos la anticoagulación y la incidencia de episodios cardiovasculares mayores en pacientes con FA ingresados por la COVID-19. Métodos: Retrospectivamente, se identificaron todos los pacientes con FA ingresados por la COVID-19 entre marzo y mayo de 2020, en 9 hospitales españoles. Se seleccionó un grupo control de pacientes ingresados consecutivamente por la COVID-19 sin FA. Se compararon las características basales, incidencia de hemorragias mayores, episodios trombóticos y mortalidad. Para reducir potenciales factores de confusión se realizó un emparejamiento por puntuación de propensión, así como un análisis multivariante para predecir hemorragia mayor y mortalidad. Resultados: Se incluyeron 305 pacientes con FA ingresados por la COVID-19. Tras el emparejamiento por puntuación de propensión, 151 pacientes con FA fueron emparejados con 151 controles. Durante el ingreso, la heparina de bajo peso molecular fue el principal anticoagulante y la incidencia de hemorragia mayor y mortalidad fue mayor en el grupo de FA (16[10,6%] vs. 3[2%], p = 0,003; 52[34,4%] vs. 35[23,2%], p = 0,03, respectivamente). El análisis multivariante demostró la presencia de FA como predictor independiente de sangrados y mortalidad intrahospitalaria en los pacientes con la COVID-19. En el grupo de FA, un segundo análisis multivariante identificó valores elevados de dímero-D como predictor independiente de hemorragia mayor intrahospitalaria. Conclusiones: Los pacientes con FA ingresados por la COVID-19 representan una población de alto riesgo de sangrado y mortalidad durante el ingreso. Parece recomendable individualizar la anticoagulación durante el ingreso, considerando el riesgo específico de sangrado y trombosis.

7.
Medicina clinica (English ed.) ; 158(12):569-575, 2022.
Article in English | EuropePMC | ID: covidwho-1905241

ABSTRACT

Introduction and purpose Atrial fibrillation (AF) is common in patients admitted with severe COVID-19. However, there is limited data about the management of chronic anticoagulation therapy in these patients. We assessed the anticoagulation and incidence of major cardiovascular events in hospitalized patients with AF and COVID-19. Methods We retrospectively investigated all consecutive patients with AF admitted with COVID-19 between March and May 2020 in 9 Spanish hospitals. We selected a control group of non-AF patients consecutively admitted with COVID-19. We compared baseline characteristics, incidence of major bleeding, thrombotic events and mortality. We used propensity score matching (PSM) to minimize potential confounding variables, as well as a multivariate analysis to predict major bleeding and death. Results 305 patients admitted with AF and COVID-19 were included. After PSM, 151 AF patients were matched with 151 control group patients. During admission, low-molecular-weight heparin was the principal anticoagulant and the incidence of major bleeding and mortality were higher in the AF group [16 (10.6%) vs 3 (2%), p = 0.003;52 (34.4%) vs 35 (23.2%), p = 0.03, respectively]. The multivariate analysis showed the presence of AF as independent predictor of in-hospital major bleeding and mortality in COVID-19 patients. In AF group, a secondary multivariate analysis identified high levels of D-dimer as independent predictor of in-hospital major bleeding. Conclusions AF patients admitted with COVID-19 represent a population at high risk for bleeding and mortality during admission. It seems advisable to individualize anticoagulation therapy during admission, considering patient specific bleeding and thrombotic risk.

8.
An Pediatr (Engl Ed) ; 93(3): 202-205, 2020 Sep.
Article in English | MEDLINE | ID: covidwho-1767883
9.
J Biomol Struct Dyn ; 41(5): 1603-1616, 2023 03.
Article in English | MEDLINE | ID: covidwho-1612284

ABSTRACT

COVID-19, a disease caused by the SARS-CoV-2 virus, is responsible for a pandemic since March 2020 and it has no cure. Therefore, herein, different theoretical methods were used to obtain potential candidates from herbal compounds to inhibit the SARS-CoV-2 main protease (Mpro). Initially, the 16 best-scored compounds were selected from a library containing 4066 ligands using virtual screening by molecular docking. Among them, six molecules (physalin B 5,6-epoxide (PHY), methyl amentoflavone (MAM), withaphysalin C (WPC), daphnoline or trilobamine (TRI), cepharanoline (CEP) and tetrandrine (TET)) were selected based on Lipinski's rule and ADMET analysis as criteria. These compounds complexed with the Mpro were submitted to triplicate 100 ns molecular dynamics simulations. RMSD, RMSF, and radius of gyration results show that the overall protein structure is preserved along the simulation time. The average ΔGbinding values, calculated by the MM/PBSA method, were -41.7, -55.8, -45.2, -38.7, -49.3, and -57.9 kcal/mol for the PHY-Mpro, MAM-Mpro, WPC-Mpro, CEP-Mpro, TRI-Mpro, and TET-Mpro complexes, respectively. Pairwise decomposition analyses revealed that the binding pocket is formed by His41-Val42, Met165-Glu166-Leu167, Asp187, and Gln189. The PLS regression model generated by QSPR analysis indicated that non-polar and polar groups with the presence of hydrogen bond acceptors play an important role in the herbal compounds-Mpro interactions. Overall, we found six potential candidates to inhibit the SARS-CoV-2 Mpro and highlighted key residues from the binding pocket that can be used for future drug design. Communicated by Ramaswamy H. Sarma.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Phytotherapy , Protease Inhibitors , SARS-CoV-2 , Humans , COVID-19/therapy , Molecular Docking Simulation , Molecular Dynamics Simulation , Protease Inhibitors/pharmacology , SARS-CoV-2/drug effects , Phytotherapy/methods
10.
Med Clin (Barc) ; 158(12): 569-575, 2022 06 24.
Article in English, Spanish | MEDLINE | ID: covidwho-1309340

ABSTRACT

INTRODUCTION AND PURPOSE: Atrial fibrillation (AF) is common in patients admitted with severe COVID-19. However, there is limited data about the management of chronic anticoagulation therapy in these patients. We assessed the anticoagulation and incidence of major cardiovascular events in hospitalized patients with AF and COVID-19. METHODS: We retrospectively investigated all consecutive patients with AF admitted with COVID-19 between March and May 2020 in 9 Spanish hospitals. We selected a control group of non-AF patients consecutively admitted with COVID-19. We compared baseline characteristics, incidence of major bleeding, thrombotic events and mortality. We used propensity score matching (PSM) to minimize potential confounding variables, as well as a multivariate analysis to predict major bleeding and death. RESULTS: 305 patients admitted with AF and COVID-19 were included. After PSM, 151 AF patients were matched with 151 control group patients. During admission, low-molecular-weight heparin was the principal anticoagulant and the incidence of major bleeding and mortality were higher in the AF group [16 (10.6%) vs 3 (2%), p=0.003; 52 (34.4%) vs 35 (23.2%), p=0.03, respectively]. The multivariate analysis showed the presence of AF as independent predictor of in-hospital major bleeding and mortality in COVID-19 patients. In AF group, a secondary multivariate analysis identified high levels of D-dimer as independent predictor of in-hospital major bleeding. CONCLUSIONS: AF patients admitted with COVID-19 represent a population at high risk for bleeding and mortality during admission. It seems advisable to individualize anticoagulation therapy during admission, considering patient specific bleeding and thrombotic risk.


Subject(s)
Atrial Fibrillation , COVID-19 , Thrombosis , Anticoagulants/adverse effects , Atrial Fibrillation/complications , Atrial Fibrillation/drug therapy , Atrial Fibrillation/epidemiology , COVID-19/complications , Hemorrhage/chemically induced , Hemorrhage/epidemiology , Humans , Prevalence , Retrospective Studies , Risk Factors , SARS-CoV-2
11.
The FASEB Journal ; 35(S1), 2021.
Article in English | Wiley | ID: covidwho-1233940

ABSTRACT

The coronavirus disease-2019 (COVID-19) has led to one of the major outbreaks in modern history. The etiological agent of COVID-19 is the novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). SARS-CoV-2 infects the respiratory system's epithelial cells via the angiotensin-converting enzyme 2 (ACE2) receptor, causing from non or mild symptoms to severe viral pneumonia, and in some cases, death. Based on evolving clinical evidence, it is now unfitting to label SARS-CoV-2 as just a respiratory virus, as lately, various reports substantiate its pathogenicity in other organs of the body, including the central nervous system (CNS). Despite conflicting results regarding ACE2 expression in brain tissue, the latest data show that ACE2 is robustly expressed by CNS cells, suggesting that CNS is a potential target for SARS-CoV-2 infection. Astrocytes are the most abundant glial cell type within the CNS and play a role in axon guidance and synaptic support, control the blood-brain barrier flow, and regulate neuroinflammation. To test if SARS-CoV-2 can enter into the CNS via astrocytes and then elicit adverse neurological effects, we have initiated studies to test if astrocytes express ACE2 and whether SARS-CoV-2 can infect human astrocytes via this receptor. We have found that astrocytes robustly express ACE2 at the mRNA and protein levels. We are currently testing if astrocytes are susceptible to SARS-CoV-2 infection, which could subsequently lead to neuronal damage and possibly account for some of the neurological symptoms observed in COVID-19 patients. We hope to contribute to gain insights into the mechanisms of SARS-CoV-2 infection in the CNS and its potential physiological effects with this work.

12.
Arq. bras. cardiol ; 115(3):558-568, 2020.
Article in English | LILACS (Americas) | ID: grc-742271

ABSTRACT

Introdução A pandemia global da doença de coronavírus 2019 (COVID-19) causada pelo novo vírus de síndrome respiratória aguda grave coronavírus 2 (SARS-CoV-2) começou em Wuhan, China, em dezembro de 2019 e afetou mais de 4,4 milhões de pessoas em todo o mundo, com 302.169 mortes até o dia 16 de maio de 2020.1 Embora os sintomas respiratórios sejam a apresentação mais comum de COVID-19, o envolvimento cardíaco é uma característica proeminente dessa doença, ocorrendo em 20% a 30% dos pacientes hospitalizados e contribuindo para 40% dos óbitos.2-4 O envolvimento cardíaco relacionado à COVID-19 tem sido documentado por elevações em biomarcadores cardíacos e frequentemente apresenta alterações no segmento ST-T no eletrocardiograma (ECG) de 12 derivações, motivo pelo qual a equipe do laboratório de cateterismo é frequentemente ativada. Além disso, as atividades do laboratório de cateterismo devem continuar no atendimento a pacientes não COVID-19 que apresentam síndrome coronariana aguda (SCA) verdadeira, infarto do miocárdio com supradesnivelamento do segmento ST (IAMCSST) e doença cardíaca isquêmica estável muito sintomática. Devido à escalada no número de casos de COVID-19 na cidade de São Paulo, epicentro da doença no Brasil, reformularam-se a logística e as práticas no laboratório de cateterismo cardíaco do Instituto Dante Pazzanese de Cardiologia, que entraram em vigor em abril de 2020 e continuarão durante o período da pandemia. Os objetivos são fornecer atendimento otimizado à população que necessita de procedimentos cardíacos invasivos durante a pandemia, com a proteção adequada aos profissionais de saúde (PS), pacientes e seus familiares. Os protocolos aqui descritos representam os esforços multidisciplinares e dinâmicos do Departamento de Cardiologia Invasiva do Instituto Dante Pazzanese de Cardiologia validados pelo Comitê de Controle de Infecção da instituição. Essas práticas estão sujeitas a alterações em função do estado epidemiológico local, a fase da epidemia e a disponibilidade de equipamento de proteção individual (EPI). Estes protocolos podem não se aplicar a outras localidades sem casos (ou casos esporádicos) de COVID-19 ou a serviços que atendem diferentes perfis populacionais com logísticas e disponibilidade de EPI diversas.

13.
Sci Rep ; 10(1): 21417, 2020 12 08.
Article in English | MEDLINE | ID: covidwho-967294

ABSTRACT

SARS-CoV-2 is a rapidly evolving pandemic causing great morbimortality. Medical therapy with hydroxicloroquine, azitromycin and protease inhibitors is being empirically used, with reported data of QTc interval prolongation. Our aim is to assess QT interval behaviour in a not critically ill and not monitored cohort of patients. We evaluated admitted and ambulatory patients with COVID-19 patients with 12 lead electrocardiogram at 48 h after treatment initiation. Other clinical and analytical variables were collected. Statistical analysis was performed to assess the magnitude of the QT interval prolongation under treatment and to identify clinical, analytical and electrocardiographic risk markers of QT prolongation independent predictors. We included 219 patients (mean age of 63.6 ± 17.4 years, 48.9% were women and 16.4% were outpatients. The median baseline QTc was 416 ms (IQR 404-433), and after treatment QTc was prolonged to 423 ms (405-438) (P < 0.001), with an average increase of 1.8%. Most of the patients presented a normal QTc under treatment, with only 31 cases (14.1%) showing a QTc interval > 460 ms, and just one case with QTc > 500 ms. Advanced age, longer QTc basal at the basal ECG and lower potassium levels were independent predictors of QTc interval prolongation. Ambulatory and not critically ill patients with COVID-19 treated with hydroxychloroquine, azithromycin and/or antiretrovirals develop a significant, but not relevant, QT interval prolongation.


Subject(s)
Antiviral Agents/adverse effects , Azithromycin/adverse effects , Hydroxychloroquine/adverse effects , Long QT Syndrome/chemically induced , Protease Inhibitors/adverse effects , Ventricular Fibrillation/chemically induced , Adult , Aged , Aged, 80 and over , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , Antimalarials/adverse effects , Antimalarials/therapeutic use , Antiviral Agents/therapeutic use , Azithromycin/therapeutic use , Critical Illness , Drug Therapy, Combination , Electrocardiography , Female , Humans , Hydroxychloroquine/therapeutic use , Male , Middle Aged , Potassium/blood , Protease Inhibitors/therapeutic use , Risk Factors , SARS-CoV-2/drug effects , Young Adult , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL