Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Clin Chim Acta ; 544: 117357, 2023 04 01.
Article in English | MEDLINE | ID: covidwho-2315526
2.
N Engl J Med ; 384(22): 2092-2101, 2021 06 03.
Article in English | MEDLINE | ID: covidwho-2283980

ABSTRACT

BACKGROUND: Several cases of unusual thrombotic events and thrombocytopenia have developed after vaccination with the recombinant adenoviral vector encoding the spike protein antigen of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (ChAdOx1 nCov-19, AstraZeneca). More data were needed on the pathogenesis of this unusual clotting disorder. METHODS: We assessed the clinical and laboratory features of 11 patients in Germany and Austria in whom thrombosis or thrombocytopenia had developed after vaccination with ChAdOx1 nCov-19. We used a standard enzyme-linked immunosorbent assay to detect platelet factor 4 (PF4)-heparin antibodies and a modified (PF4-enhanced) platelet-activation test to detect platelet-activating antibodies under various reaction conditions. Included in this testing were samples from patients who had blood samples referred for investigation of vaccine-associated thrombotic events, with 28 testing positive on a screening PF4-heparin immunoassay. RESULTS: Of the 11 original patients, 9 were women, with a median age of 36 years (range, 22 to 49). Beginning 5 to 16 days after vaccination, the patients presented with one or more thrombotic events, with the exception of 1 patient, who presented with fatal intracranial hemorrhage. Of the patients with one or more thrombotic events, 9 had cerebral venous thrombosis, 3 had splanchnic-vein thrombosis, 3 had pulmonary embolism, and 4 had other thromboses; of these patients, 6 died. Five patients had disseminated intravascular coagulation. None of the patients had received heparin before symptom onset. All 28 patients who tested positive for antibodies against PF4-heparin tested positive on the platelet-activation assay in the presence of PF4 independent of heparin. Platelet activation was inhibited by high levels of heparin, Fc receptor-blocking monoclonal antibody, and immune globulin (10 mg per milliliter). Additional studies with PF4 or PF4-heparin affinity purified antibodies in 2 patients confirmed PF4-dependent platelet activation. CONCLUSIONS: Vaccination with ChAdOx1 nCov-19 can result in the rare development of immune thrombotic thrombocytopenia mediated by platelet-activating antibodies against PF4, which clinically mimics autoimmune heparin-induced thrombocytopenia. (Funded by the German Research Foundation.).


Subject(s)
Autoantibodies/blood , COVID-19 Vaccines/adverse effects , Platelet Factor 4/immunology , Thrombocytopenia/etiology , Thrombosis/etiology , Adult , Autoimmune Diseases/etiology , Blood Chemical Analysis , ChAdOx1 nCoV-19 , Disseminated Intravascular Coagulation/etiology , Enzyme-Linked Immunosorbent Assay , Fatal Outcome , Female , Humans , Intracranial Hemorrhages/etiology , Male , Middle Aged , Platelet Activation , Thrombocytopenia/immunology , Thrombosis/immunology , Young Adult
3.
Stud Hist Philos Sci ; 92: 45-55, 2022 04.
Article in English | MEDLINE | ID: covidwho-1665463

ABSTRACT

Twentieth-century medicine saw the remarkable rise of complex machines and infrastructures to process blood for medical purposes, such as transfusion, dialysis, and cardiac surgery. Instead of attributing these developments to technological ingenuity, this article argues for the primacy of material encounters as a promising focal point of medical historiography. In fact, blood's special properties consistently clashed with most materials used in medical practice, provoking a series of material exchanges. Drawing on a combination of epistemological and network approaches, three exemplary cases are presented to examine blood's encounters with plastics, plant and animal extracts: William M. Bayliss's (1860-1926) injections of dissolved gum acacia to expand diminished blood volume; Charles H. Best's (1899-1978) production of the anticoagulant heparin from animal organs; and the preservation of fragile blood cells by silicone coatings inside of John H. Gibbon Jr.'s (1903-1973) heart-lung machine. The case studies demonstrate how the complementarity of blood and these materials produced hybridizations between medicine and a range of industrial branches, from colonial forestry and meatpacking to commercial chemistry. In this light, the paper concludes by discussing the dependencies of today's healthcare environments on globally distributed, capitalistically appropriated resources in the face of crises like the COVID-19 pandemic.


Subject(s)
Blood , Medicine , Plastics , Animals , Blood Chemical Analysis , Blood Physiological Phenomena , History, 20th Century , Humans , Plant Extracts , Plastics/chemistry
4.
Dis Markers ; 2022: 5106342, 2022.
Article in English | MEDLINE | ID: covidwho-1650406

ABSTRACT

OBJECTIVE: An outbreak of coronavirus disease-19 (COVID-19) began in December 2019 and spread globally, overwhelming the entire world. COVID-19 is a public health emergency of international concern. Due to its high morbidity and mortality rate, recognition of its risk and prognostic factors is important. We aimed to understand the relationship between metabolic and endocrine parameters and the prognosis of COVID-19. METHODS AND MATERIALS: This was a cross-sectional clinical study. A total of 70 patients with severe COVID-19 were enrolled. Laboratory results at the first admission time (including complete blood count, C-reactive protein, lactate dehydrogenase, blood glucose, calcium, phosphate, albumin, creatinine, magnesium, lipid profiles, liver enzymes, thyroid hormones, cortisol, and vitamin D) and outcome data were recorded. We divided patients into (1) intensive care unit- (ICU-) admitted and non-ICU-admitted and (2) survivors and nonsurvivors for estimation of severity and prognosis. We determined the risk factors associated with critical illness and poor prognosis. RESULTS: Patients with higher white blood cell (WBC) count and phosphate levels had significantly higher ICU admission rates. According to univariate analysis, serum levels of T3, phosphate, and WBC as well as the duration of hospitalization were associated with mortality. Multivariate analysis revealed that only WBC and duration of hospitalization were independent predictors for mortality rate in COVID-19 patients. CONCLUSION: Our findings suggest that longer duration of hospitalization and higher WBC count are associated with poor outcomes in patients with COVID-19.


Subject(s)
COVID-19/etiology , COVID-19/mortality , Endocrine System/metabolism , Leukocyte Count , Phosphates/blood , Aged , Biomarkers , Blood Chemical Analysis , Cross-Sectional Studies , Endocrine System/virology , Female , Humans , Intensive Care Units , Length of Stay , Male , Middle Aged , Mortality , Multivariate Analysis , Prognosis , Severity of Illness Index , Vitamin D/blood
5.
J Med Virol ; 94(1): 357-365, 2022 01.
Article in English | MEDLINE | ID: covidwho-1544349

ABSTRACT

COVID-19 is a serious respiratory disease. The ever-increasing number of cases is causing heavier loads on the health service system. Using 38 blood test indicators on the first day of admission for the 422 patients diagnosed with COVID-19 (from January 2020 to June 2021) to construct different machine learning (ML) models to classify patients into either mild or severe cases of COVID-19. All models show good performance in the classification between COVID-19 patients into mild and severe disease. The area under the curve (AUC) of the random forest model is 0.89, the AUC of the naive Bayes model is 0.90, the AUC of the support vector machine model is 0.86, and the AUC of the KNN model is 0.78, the AUC of the Logistic regression model is 0.84, and the AUC of the artificial neural network model is 0.87, among which the naive Bayes model has the best performance. Different ML models can classify patients into mild and severe cases based on 38 blood test indicators taken on the first day of admission for patients diagnosed with COVID-19.


Subject(s)
Blood Chemical Analysis , COVID-19/classification , Neural Networks, Computer , Severity of Illness Index , Support Vector Machine , Area Under Curve , COVID-19/blood , COVID-19/diagnosis , Hematologic Tests , Humans , Logistic Models , SARS-CoV-2
6.
J Clin Lab Anal ; 35(11): e24011, 2021 Nov.
Article in English | MEDLINE | ID: covidwho-1525448

ABSTRACT

BACKGROUND: Interleukin 6 assays are useful in early detection of infections and risk stratification of critically ill patients, so an assay with a short turnaround-time and near-patient use is preferred. This study evaluated the performance of a new interleukin 6 assay, Pylon IL-6 assay, and explored its potential use in near-patient settings. METHODS: We carried out imprecision, linearity and comparison studies using serum and plasma samples according to CLSI EP guidelines. The stability of whole blood samples during storage was assessed. Furthermore, whole blood samples from pediatric patients with suspected infection were measured to evaluate the assay's diagnostic performance. RESULTS: The within-run CVs and total CVs of Pylon IL-6 assay were determined as 1.8% and 3.0% at 159.3 pg/ml and 3.5% and 4.7% at 8009.9 pg/ml, respectively. The method showed linearity between 1.5 and 42,854 pg/ml. The results of serum samples measured by Pylon assays correlated to those measured by Roche assays, as well as to those of matched whole blood samples measured by Pylon assays. IL-6 in whole blood was found stable for ~8 h at room temperature. Pylon IL-6 results of whole blood samples from 179 pediatric patients with suspected infection showed an AUC of 0.842 in diagnosis of bacterial infection. The turnaround time of Pylon IL-6 assay was only 1 h when using whole blood samples. CONCLUSION: The new assay demonstrated performance comparable to those performed on clinical laboratory instruments and can be used in near-patient settings with whole blood to reduce turnaround times.


Subject(s)
Blood Chemical Analysis , Immunoassay , Interleukin-6/blood , Blood Chemical Analysis/methods , Blood Chemical Analysis/standards , Child , Child, Preschool , Female , Humans , Immunoassay/methods , Immunoassay/standards , Infant , Limit of Detection , Linear Models , Male , Reproducibility of Results
7.
J Cell Mol Med ; 25(24): 11212-11220, 2021 12.
Article in English | MEDLINE | ID: covidwho-1511334

ABSTRACT

This study aims to evaluate the effect of non-alcoholic fatty liver disease (NAFLD) on the susceptibility and consequences of coronavirus disease 2019 (COVID-19). We retrospectively collected data from 218 adult COVID-19 patients who showed no evidence of excessive alcohol consumption and underwent abdominal ultrasound examinations. Of these patients, 39.4% patients had been diagnosed with NAFLD, which indicates a much higher prevalence of NAFLD than that reported in the general population. Significantly elevated white blood cell count (p = 0.008), alanine aminotransferase (p = 0.000), aspartate aminotransferase (p = 0.006) and C reactive protein (p = 0.012) were found in the patients with NAFLD. These patients also had significantly higher proportions of hypertension (p = 0.006) and diabetes (p = 0.049) than the non-NAFLD cases. No significant differences existed in the severity, mortality, viral shedding time and length of hospital stay between patients with or without NAFLD in the sample population. However, subgroup analyses found that in patients with normal body mass index (BMI), NAFLD sufferers were more likely to experience a severe event (30.0% vs 11.5%, p = 0.021). Kaplan-Meier curve (log-rank p = 0.017) and Cox regression (HR = 3.26, 95% CI: 1.17-9.04, p = 0.023) analyses confirmed that before and after adjusting for gender, age and comorbidities, NAFLD patients with normal BMI had a higher incidence of suffering severe events. People with NAFLD may have a higher proportion of COVID-19. NAFLD may be correlated with the severity of COVID-19 patients in the normal BMI group.


Subject(s)
COVID-19/etiology , Non-alcoholic Fatty Liver Disease/etiology , Adult , Aged , Aged, 80 and over , Blood Chemical Analysis , Body Mass Index , COVID-19/epidemiology , COVID-19/therapy , Comorbidity , Disease Susceptibility , Female , Humans , Incidence , Kaplan-Meier Estimate , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/epidemiology , Prevalence , Retrospective Studies , Virus Shedding , Young Adult
8.
J Mol Med (Berl) ; 100(2): 285-301, 2022 02.
Article in English | MEDLINE | ID: covidwho-1505851

ABSTRACT

The risk of severe COVID-19 increases with age as older patients are at highest risk. Thus, there is an urgent need to identify how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) interacts with blood components during aging. We investigated the whole blood transcriptome from the Genotype-Tissue Expression (GTEx) database to explore differentially expressed genes (DEGs) translated into proteins interacting with viral proteins during aging. From 22 DEGs in aged blood, FASLG, CTSW, CTSE, VCAM1, and BAG3 were associated with immune response, inflammation, cell component and adhesion, and platelet activation/aggregation. Males and females older than 50 years old overexpress FASLG, possibly inducing a hyperinflammatory cascade. The expression of cathepsins (CTSW and CTSE) and the anti-apoptotic co-chaperone molecule BAG3 also increased throughout aging in both genders. By exploring single-cell RNA-sequencing data from peripheral blood of SARS-CoV-2-infected patients, we found FASLG and CTSW expressed in natural killer cells and CD8 + T lymphocytes, whereas BAG3 was expressed mainly in CD4 + T cells, naive T cells, and CD14 + monocytes. In addition, T cell exhaustion was associated with increased expression of CCL4L2 and DUSP4 over blood aging. LAG3, PDCD1, TIGIT, VCAM1, HLA-DRA, and TOX also increased in individuals aged 60-69 years old; conversely, the RGS2 gene decreased with aging. We further identified a distinct gene expression profile associated with type I interferon signaling following blood aging. These results revealed changes in blood molecules potentially related to SARS-CoV-2 infection throughout aging, emphasizing them as therapeutic candidates for aggressive clinical manifestation of COVID-19. KEY MESSAGES: • Prediction of host-viral interactions in the whole blood transcriptome during aging. • Expression levels of FASLG, CTSW, CTSE, VCAM1, and BAG3 increase in aged blood. • Blood interactome reveals targets involved with immune response, inflammation, and blood clots. • SARS-CoV-2-infected patients with high viral load showed FASLG overexpression. • Gene expression profile associated with T cell exhaustion and type I interferon signaling were affected with blood aging.


Subject(s)
Aging/blood , Blood Proteins/analysis , COVID-19/genetics , SARS-CoV-2/pathogenicity , Transcriptome , Adult , Aged , Aging/genetics , Blood/metabolism , Blood Chemical Analysis , Blood Proteins/genetics , Blood Proteins/metabolism , Blood Vessels/metabolism , Blood Vessels/virology , COVID-19/blood , COVID-19/immunology , COVID-19/physiopathology , Cardiovascular Physiological Phenomena/genetics , Cardiovascular System/metabolism , Cardiovascular System/virology , Cohort Studies , Female , Genetic Association Studies , Humans , Immunity, Innate/genetics , Male , Middle Aged , Young Adult
9.
Sci Rep ; 11(1): 21136, 2021 10 27.
Article in English | MEDLINE | ID: covidwho-1493228

ABSTRACT

The COVID-19 pandemic is impressively challenging the healthcare system. Several prognostic models have been validated but few of them are implemented in daily practice. The objective of the study was to validate a machine-learning risk prediction model using easy-to-obtain parameters to help to identify patients with COVID-19 who are at higher risk of death. The training cohort included all patients admitted to Fondazione Policlinico Gemelli with COVID-19 from March 5, 2020, to November 5, 2020. Afterward, the model was tested on all patients admitted to the same hospital with COVID-19 from November 6, 2020, to February 5, 2021. The primary outcome was in-hospital case-fatality risk. The out-of-sample performance of the model was estimated from the training set in terms of Area under the Receiving Operator Curve (AUROC) and classification matrix statistics by averaging the results of fivefold cross validation repeated 3-times and comparing the results with those obtained on the test set. An explanation analysis of the model, based on the SHapley Additive exPlanations (SHAP), is also presented. To assess the subsequent time evolution, the change in paO2/FiO2 (P/F) at 48 h after the baseline measurement was plotted against its baseline value. Among the 921 patients included in the training cohort, 120 died (13%). Variables selected for the model were age, platelet count, SpO2, blood urea nitrogen (BUN), hemoglobin, C-reactive protein, neutrophil count, and sodium. The results of the fivefold cross-validation repeated 3-times gave AUROC of 0.87, and statistics of the classification matrix to the Youden index as follows: sensitivity 0.840, specificity 0.774, negative predictive value 0.971. Then, the model was tested on a new population (n = 1463) in which the case-fatality rate was 22.6%. The test model showed AUROC 0.818, sensitivity 0.813, specificity 0.650, negative predictive value 0.922. Considering the first quartile of the predicted risk score (low-risk score group), the case-fatality rate was 1.6%, 17.8% in the second and third quartile (high-risk score group) and 53.5% in the fourth quartile (very high-risk score group). The three risk score groups showed good discrimination for the P/F value at admission, and a positive correlation was found for the low-risk class to P/F at 48 h after admission (adjusted R-squared = 0.48). We developed a predictive model of death for people with SARS-CoV-2 infection by including only easy-to-obtain variables (abnormal blood count, BUN, C-reactive protein, sodium and lower SpO2). It demonstrated good accuracy and high power of discrimination. The simplicity of the model makes the risk prediction applicable for patients in the Emergency Department, or during hospitalization. Although it is reasonable to assume that the model is also applicable in not-hospitalized persons, only appropriate studies can assess the accuracy of the model also for persons at home.


Subject(s)
COVID-19/mortality , Machine Learning , Pandemics , SARS-CoV-2 , Aged , Aged, 80 and over , Blood Cell Count , Blood Chemical Analysis , COVID-19/blood , Cohort Studies , Female , Hospital Mortality , Humans , Male , Middle Aged , Models, Statistical , Multivariate Analysis , Oxygen/blood , Pandemics/statistics & numerical data , ROC Curve , Risk Factors , Rome/epidemiology
10.
Microbiol Spectr ; 9(2): e0054921, 2021 10 31.
Article in English | MEDLINE | ID: covidwho-1381170

ABSTRACT

In one year of the coronavirus disease 2019 (COVID-19) pandemic, many studies have described the different metabolic changes occurring in COVID-19 patients, linking these alterations to the disease severity. However, a complete metabolic signature of the most severe cases, especially those with a fatal outcome, is still missing. Our study retrospectively analyzes the metabolome profiles of 75 COVID-19 patients with moderate and severe symptoms admitted to Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico (Lombardy Region, Italy) following SARS-CoV-2 infection between March and April 2020. Italy was the first Western country to experience COVID-19, and the Lombardy Region was the epicenter of the Italian COVID-19 pandemic. This cohort shows a higher mortality rate compared to others; therefore, it represents a unique opportunity to investigate the underlying metabolic profiles of the first COVID-19 patients in Italy and to identify the potential biomarkers related to the disease prognosis and fatal outcome. IMPORTANCE Understanding the metabolic alterations occurring during an infection is a key element for identifying potential indicators of the disease prognosis, which are fundamental for developing efficient diagnostic tools and offering the best therapeutic treatment to the patient. Here, exploiting high-throughput metabolomics data, we identified the first metabolic profile associated with a fatal outcome, not correlated with preexisting clinical conditions or the oxygen demand at the moment of diagnosis. Overall, our results contribute to a better understanding of COVID-19-related metabolic disruption and may represent a useful starting point for the identification of independent prognostic factors to be employed in therapeutic practice.


Subject(s)
Blood Chemical Analysis , COVID-19/epidemiology , COVID-19/mortality , Energy Metabolism/physiology , Metabolome/physiology , Aged , Aged, 80 and over , Biomarkers/blood , Comorbidity , Female , Humans , Italy/epidemiology , Male , Middle Aged , Prognosis , Retrospective Studies , SARS-CoV-2
11.
Ann Emerg Med ; 78(4): 511-514, 2021 10.
Article in English | MEDLINE | ID: covidwho-1293546

ABSTRACT

Vaccine-induced thrombotic thrombocytopenia is a newly described disease process in the setting of expanding access to COVID-19 vaccination. The United States Centers for Disease Control and Prevention recommends treatment with an alternative to heparin in patients suspected of having vaccine-induced thrombotic thrombocytopenia. At this time there have been no reported outcomes from the treatment of vaccine-induced thrombotic thrombocytopenia with bivalirudin as a heparin alternative. We describe the early outcomes from the treatment of vaccine-induced thrombotic thrombocytopenia with bivalirudin as a heparin alternative. A 40-year-old Caucasian woman was found to have thrombocytopenia, cerebral venous sinus thrombosis, and pulmonary embolism following vaccination for COVID-19 with Ad26.COV2.S. She exhibited a steady rise in platelet count: 20×109/L at hospital day 0, 115×109/L at discharge on hospital day 6, and 182×109/L on outpatient follow-up on day 9. While the patient exhibited a transient drop in hemoglobin, there was no clinical evidence of bleeding. This patient did not demonstrate any clinical sequelae of thrombosis, and she reported resolution of her headache. Vaccination with Ad26.COV2.S appears to be associated with a small but significant risk for thrombotic thrombocytopenia within 13 days of receipt. The Centers for Disease Control and Prevention guidance to consider an alternative to heparin was not accompanied by specifically recommended alternatives. A single patient treated with bivalirudin for suspected vaccine-induced thrombotic thrombocytopenia subsequently experienced symptom improvement and a rise in platelet count and did not demonstrate any immediate negative outcomes. A provider may consider bivalirudin as an alternative to heparin in patients with suspected vaccine-induced thrombotic thrombocytopenia following Ad26.COV2.S vaccination, pending more definitive research.


Subject(s)
COVID-19 Vaccines/adverse effects , Fibrinolytic Agents/therapeutic use , Peptide Fragments/therapeutic use , Sinus Thrombosis, Intracranial/drug therapy , Thrombocytopenia/drug therapy , Ad26COVS1 , Adult , Blood Chemical Analysis , Blood Physiological Phenomena , COVID-19/prevention & control , Female , Hirudins , Humans , Pulmonary Embolism/drug therapy , Pulmonary Embolism/etiology , Recombinant Proteins/therapeutic use , Sinus Thrombosis, Intracranial/etiology , Thrombocytopenia/etiology
13.
Ann Clin Biochem ; 58(5): 411-421, 2021 09.
Article in English | MEDLINE | ID: covidwho-1181016

ABSTRACT

BACKGROUND: The COVID-19 pandemic has drastically changed the delivery of secondary care services. Self-collection of capillary blood at home can facilitate the monitoring of patients with chronic disease to support virtual clinics while mitigating the risk of SARS-CoV-2 infection and transmission. OBJECTIVE: To investigate the comparability of whole blood capillary and plasma venous samples for 15 routinely used biochemical analytes and to develop and pilot a user-friendly home-collection kit to support virtual outpatient clinical services. METHODS: To investigate the comparability of whole blood capillary and plasma venous samples for 15 routinely requested biochemical analytes, simultaneous samples of venous and capillary blood were collected in EDTA and lithium-heparin plasma separation tubes that were of 4-6 mL and 400-600 µL draw volume, respectively. Venous samples were analysed within 4 h of collection while capillary samples were kept at ambient temperature for three days until centrifugation and analysis. Analyte results that were comparable between the matrices were then piloted in a feasibility study in three outpatient clinical services. RESULTS: HbA1c, lipid profile and liver function tests were considered comparable and piloted in the patient feasibility study. The home-collect kit demonstrated good patient usability. CONCLUSION: Home collection of capillary blood could be a clinically-useful tool to deliver virtual care to patients with chronic disease.


Subject(s)
Blood Chemical Analysis/methods , Blood Specimen Collection/methods , COVID-19/blood , Pandemics , SARS-CoV-2 , Adult , Blood Chemical Analysis/instrumentation , Blood Specimen Collection/instrumentation , Capillary Tubing , Feasibility Studies , Female , Humans , London , Male , Middle Aged , Phlebotomy/instrumentation , Phlebotomy/methods , Pilot Projects , Remote Consultation , Self Care/instrumentation , Self Care/methods , Surveys and Questionnaires
14.
N Engl J Med ; 384(22): 2124-2130, 2021 06 03.
Article in English | MEDLINE | ID: covidwho-1174740

ABSTRACT

We report findings in five patients who presented with venous thrombosis and thrombocytopenia 7 to 10 days after receiving the first dose of the ChAdOx1 nCoV-19 adenoviral vector vaccine against coronavirus disease 2019 (Covid-19). The patients were health care workers who were 32 to 54 years of age. All the patients had high levels of antibodies to platelet factor 4-polyanion complexes; however, they had had no previous exposure to heparin. Because the five cases occurred in a population of more than 130,000 vaccinated persons, we propose that they represent a rare vaccine-related variant of spontaneous heparin-induced thrombocytopenia that we refer to as vaccine-induced immune thrombotic thrombocytopenia.


Subject(s)
Autoantibodies/blood , COVID-19 Vaccines/adverse effects , Platelet Factor 4/immunology , Thrombocytopenia/etiology , Thrombosis/etiology , Adult , Autoimmune Diseases/etiology , Blood Chemical Analysis , ChAdOx1 nCoV-19 , Enzyme-Linked Immunosorbent Assay , Fatal Outcome , Female , Humans , Male , Middle Aged , Platelet Aggregation , Platelet Count
15.
Int J Mol Sci ; 22(5)2021 Mar 09.
Article in English | MEDLINE | ID: covidwho-1134169

ABSTRACT

Fibrinolysis is an important process in hemostasis responsible for dissolving the clot during wound healing. Plasmin is a central enzyme in this process via its capacity to cleave fibrin. The kinetics of plasmin generation (PG) and inhibition during fibrinolysis have been poorly understood until the recent development of assays to quantify these metrics. The assessment of plasmin kinetics allows for the identification of fibrinolytic dysfunction and better understanding of the relationships between abnormal fibrin dissolution and disease pathogenesis. Additionally, direct measurement of the inhibition of PG by antifibrinolytic medications, such as tranexamic acid, can be a useful tool to assess the risks and effectiveness of antifibrinolytic therapy in hemorrhagic diseases. This review provides an overview of available PG assays to directly measure the kinetics of plasmin formation and inhibition in human and mouse plasmas and focuses on their applications in defining the role of plasmin in diseases, including angioedema, hemophilia, rare bleeding disorders, COVID-19, or diet-induced obesity. Moreover, this review introduces the PG assay as a promising clinical and research method to monitor antifibrinolytic medications and screen for genetic or acquired fibrinolytic disorders.


Subject(s)
Blood Chemical Analysis/methods , Disease , Fibrinolysin/analysis , Fibrinolysin/metabolism , Animals , Antifibrinolytic Agents/blood , Fibrin/analysis , Fibrin/chemistry , Fibrinolytic Agents/blood , Humans , Plasminogen/analysis , Plasminogen/chemistry , Plasminogen/metabolism
16.
Pharmacol Res Perspect ; 9(2): e00743, 2021 04.
Article in English | MEDLINE | ID: covidwho-1130677

ABSTRACT

Both antiviral treatment with remdesivir and hemoadsorption using a CytoSorb® adsorption device are applied in the treatment of severe COVID-19. The CytoSorb® adsorber consists of porous polymer beads that adsorb a broad range of molecules, including cytokines but also several therapeutic drugs. In this study, we evaluated whether remdesivir and its main active metabolite GS-441524 would be adsorbed by CytoSorb® . Serum containing remdesivir or GS-441524 was circulated in a custom-made system containing a CytoSorb® device. Concentrations of remdesivir and GS-441524 before and after the adsorber were analyzed by liquid chromatography-tandem mass spectrometry. Measurements of remdesivir in the outgoing tube after the adsorber indicated almost complete removal of remdesivir by the device. In the reservoir, concentration of remdesivir showed an exponential decay and was not longer detectable after 60 mins. GS-441524 showed a similar exponential decay but, unlike remdesivir, it reached an adsorption-desorption equilibrium at ~48 µg/L. Remdesivir and its main active metabolite GS-441524 are rapidly eliminated from the perfusate by the CytoSorb® adsorber device in vitro. This should be considered in patients for whom both therapies are indicated, and simultaneous application should be avoided. In general, plasma levels of therapeutic drugs should be closely monitored under concurrent CytoSorb® therapy.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , COVID-19/therapy , Hemoperfusion/instrumentation , Adenosine/analogs & derivatives , Adenosine Monophosphate/blood , Adenosine Monophosphate/pharmacokinetics , Alanine/blood , Alanine/pharmacokinetics , Blood Chemical Analysis , COVID-19/blood , Chromatography, Liquid , Combined Modality Therapy , Furans/blood , Furans/pharmacokinetics , Hemoperfusion/adverse effects , Humans , Pyrroles/blood , Pyrroles/pharmacokinetics , Tandem Mass Spectrometry , Triazines/blood , Triazines/pharmacokinetics
19.
Clin Lab ; 67(2)2021 Feb 01.
Article in English | MEDLINE | ID: covidwho-1094346

ABSTRACT

BACKGROUND: COVID-19 has recently been declared an epidemic by the WHO, and there is an urgent need for affected countries and laboratories to assess and treat people at risk of COVID-19. A heat procedure has been suggested for specimen inactivation. This study was designed to evaluate the effect of serum heating on biochemical indexes, and providing a basis for accurate detection results of the COVID-19 patients. METHODS: We collected 29 normal cases of two tubes of 5 mL whole blood. One tube was analyzed directly, and the other was analyzed after heating at 56°C 30 minutes. RESULTS: A total of 34 serum biochemical index quantitative results were obtained, 28/34 indexes were not significantly affected by the heat inactivation and remained clinically interpretable. As the thermal inactivation for these indexes showed good correlation, ALB (p = 0.04, Pearson R = 0.91, 2.6% mean increase), CysC (p = 0.03, Pearson R = 0.98, 9.9% mean increase), CO2CP (p < 0.001, Pearson R = 0.96, 13% mean decrease), they were still inter-pretable. Four biochemical indexes ALP, CK, CK-MB, and insulin were inactivated and showed significant statistical differences (p < 0.001). CONCLUSIONS: Our study showed CK, CK-MB, ALP, and insulin were sensitive to heat and will be inhibited or degrade after heating, indicating that the rapid decrease of this indexes in the COVID-19 patients may be caused by sample heat inactivation. For safety and diagnostic accuracy, we recommend the use of a point-of-care device for blood gases, electrolytes, troponin, and liver and renal function tests within a ISL 2 or above biosafety cabinet with level 3 or above biosafety laboratory practice.


Subject(s)
Blood Chemical Analysis , COVID-19 , Diagnostic Errors/prevention & control , Hot Temperature/adverse effects , SARS-CoV-2 , Virus Inactivation , Alkaline Phosphatase/blood , Blood Chemical Analysis/methods , Blood Chemical Analysis/standards , COVID-19/blood , COVID-19/diagnosis , COVID-19/virology , Creatine Kinase/blood , Female , Humans , Insulin/blood , Male , Middle Aged , Point-of-Care Systems , SARS-CoV-2/isolation & purification , SARS-CoV-2/physiology , Sensitivity and Specificity
20.
Anal Sci ; 37(9): 1301-1304, 2021 Sep 10.
Article in English | MEDLINE | ID: covidwho-1094104

ABSTRACT

An in-hospital rapid method for quantifying the serum level of favipiravir (FPV) in the pharmacological treatment of COVID-19 was developed by an appropriate combination of a solid-phase extraction treatment and a reversed-phase HPLC/UV detection system. The quantification method was well-validated and applied to measuring the serum FPV level in a clinical practice at a general hospital that accepts COVID-19 patients. Furthermore, an analysis of data from our preliminary interaction analysis revealed, for the first time, that FPV selectively forms complexes with ferric (Fe3+) and cupric (Cu2+) ions.


Subject(s)
Amides/blood , Blood Chemical Analysis/methods , COVID-19 Drug Treatment , Hospitals , Pyrazines/blood , Amides/therapeutic use , COVID-19/blood , Chromatography, High Pressure Liquid , Humans , Pyrazines/therapeutic use , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL