Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
1.
Sci Rep ; 13(1): 17992, 2023 10 21.
Article de Anglais | MEDLINE | ID: mdl-37865713

RÉSUMÉ

A20 serves as a critical brake on NF-κB-dependent inflammation. In humans, polymorphisms in or near the TNFAIP3/A20 gene have been linked to various inflammatory disorders, including systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Experimental gene knockout studies in mice have confirmed A20 as a susceptibility gene for SLE and RA. Here, we examine the significance of protein citrullination and NET formation in the autoimmune pathology of A20 mutant mice because autoimmunity directed against citrullinated antigens released by neutrophil extracellular traps (NETs) is central to the pathogenesis of RA and SLE. Furthermore, genetic variants impairing the deubiquitinase (DUB) function of A20 have been shown to contribute to autoimmune susceptibility. Our findings demonstrate that genetic disruption of A20 DUB function in A20 C103R knockin mice does not result in autoimmune pathology. Moreover, we show that PAD4 deficiency, which abolishes protein citrullination and NET formation, does not prevent the development of autoimmunity in A20 deficient mice. Collectively, these findings provide experimental confirmation that PAD4-dependent protein citrullination and NET formation do not serve as pathogenic mechanisms in the development of RA and SLE pathology in mice with A20 mutations.


Sujet(s)
Polyarthrite rhumatoïde , Pièges extracellulaires , Lupus érythémateux disséminé , Humains , Animaux , Souris , Citrullination , Polyarthrite rhumatoïde/métabolisme , Lupus érythémateux disséminé/génétique , Lupus érythémateux disséminé/métabolisme , Protéine-3 induite par le facteur de nécrose tumorale alpha/génétique , Protéine-3 induite par le facteur de nécrose tumorale alpha/métabolisme , Inflammation/métabolisme , Auto-immunité/génétique , Pièges extracellulaires/métabolisme
2.
Sci Transl Med ; 15(710): eadi0252, 2023 08 23.
Article de Anglais | MEDLINE | ID: mdl-37611083

RÉSUMÉ

Improvements in COVID-19 treatments, especially for the critically ill, require deeper understanding of the mechanisms driving disease pathology. The complement system is not only a crucial component of innate host defense but can also contribute to tissue injury. Although all complement pathways have been implicated in COVID-19 pathogenesis, the upstream drivers and downstream effects on tissue injury remain poorly defined. We demonstrate that complement activation is primarily mediated by the alternative pathway, and we provide a comprehensive atlas of the complement alterations around the time of respiratory deterioration. Proteomic and single-cell sequencing mapping across cell types and tissues reveals a division of labor between lung epithelial, stromal, and myeloid cells in complement production, in addition to liver-derived factors. We identify IL-6 and STAT1/3 signaling as an upstream driver of complement responses, linking complement dysregulation to approved COVID-19 therapies. Furthermore, an exploratory proteomic study indicates that inhibition of complement C5 decreases epithelial damage and markers of disease severity. Collectively, these results support complement dysregulation as a key druggable feature of COVID-19.


Sujet(s)
COVID-19 , Interleukine-6 , Humains , Protéomique , Protéines du système du complément , Activation du complément
3.
Semin Immunol ; 67: 101759, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-37031560

RÉSUMÉ

Despite the lack of endogenous chitin synthesis, mammalian genomes encode two enzymatically active true chitinases (chitotriosidase and acidic mammalian chitinase) and a variable number of chitinase-like proteins (CLPs) that have no enzyme activity but bind chitin. Chitinases and CLPs are prominent components of type-2 immune response-mediated respiratory diseases. However, despite extensive research into their role in allergic airway disease, there is still no agreement on whether they are mere biomarkers of disease or actual disease drivers. Functions ascribed to chitinases and CLPs include, but are not limited to host defense against chitin-containing pathogens, directly promoting inflammation, and modulating tissue remodeling and fibrosis. Here, we discuss in detail the chitin-dependent and -independent roles of chitinases and CLPs in the context of allergic airway disease, and recent advances and emerging concepts in the field that might identify opportunities for new therapies.


Sujet(s)
Asthme , Chitinase , Hypersensibilité , Animaux , Humains , Chitinase/métabolisme , Inflammation , Chitine/métabolisme , Mammifères/métabolisme
4.
Insect Mol Biol ; 32(2): 173-186, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36511188

RÉSUMÉ

RNA viruses have recently been detected in association with house dust mites, including laboratory cultures, dust samples, and mite-derived pharmaceuticals used for allergy diagnosis. This study aimed to assess the incidence of viral infection on Dermatophagoides pteronyssinus physiology and on the allergenic performance of extracts derived from its culture. Transcriptional changes between genetically identical control and virus-infected mite colonies were analysed by RNAseq with the support of a new D. pteronyssinus high-quality annotated genome (56.8 Mb, 108 scaffolds, N50 = 2.73 Mb, 96.7% BUSCO-completeness). Extracts of cultures and bodies from both colonies were compared by inspecting major allergen accumulation by enzyme-linked immunosorbent assay (ELISA), allergen-related enzymatic activities by specific assays, airway inflammation in a mouse model of allergic asthma, and binding to allergic patient's sera IgE by ImmunoCAP. Viral infection induced a significant transcriptional response, including several immunity and stress-response genes, and affected the expression of seven allergens, putative isoallergens and allergen orthologs. Major allergens were unaffected except for Der p 23 that was upregulated, increasing ELISA titers up to 29% in infected-mite extracts. By contrast, serine protease allergens Der p 3, 6 and 9 were downregulated, being trypsin and chymotrypsin enzymatic activities reduced up to 21% in extracts. None of the parameters analysed in our mouse model, nor binding to human IgE were significantly different when comparing control and infected-mite extracts. Despite the described physiological impact of viral infection on the mites, no significant consequences for the allergenicity of derived extracts or their practical use in allergy diagnosis have been detected.


Sujet(s)
Hypersensibilité , Virus à ARN , Médicaments vétérinaires , Souris , Humains , Animaux , Allergènes/analyse , Allergènes/génétique , Pyroglyphidae/métabolisme , Virus à ARN/métabolisme , Immunoglobuline E
5.
Cell Rep Med ; 3(12): 100833, 2022 12 20.
Article de Anglais | MEDLINE | ID: mdl-36459994

RÉSUMÉ

GM-CSF promotes myelopoiesis and inflammation, and GM-CSF blockade is being evaluated as a treatment for COVID-19-associated hyperinflammation. Alveolar GM-CSF is, however, required for monocytes to differentiate into alveolar macrophages (AMs) that control alveolar homeostasis. By mapping cross-species AM development to clinical lung samples, we discovered that COVID-19 is marked by defective GM-CSF-dependent AM instruction and accumulation of pro-inflammatory macrophages. In a multi-center, open-label RCT in 81 non-ventilated COVID-19 patients with respiratory failure, we found that inhalation of rhu-GM-CSF did not improve mean oxygenation parameters compared with standard treatment. However, more patients on GM-CSF had a clinical response, and GM-CSF inhalation induced higher numbers of virus-specific CD8 effector lymphocytes and class-switched B cells, without exacerbating systemic hyperinflammation. This translational proof-of-concept study provides a rationale for further testing of inhaled GM-CSF as a non-invasive treatment to improve alveolar gas exchange and simultaneously boost antiviral immunity in COVID-19. This study is registered at ClinicalTrials.gov (NCT04326920) and EudraCT (2020-001254-22).


Sujet(s)
COVID-19 , Macrophages alvéolaires , Humains , Facteur de stimulation des colonies de granulocytes et de macrophages/usage thérapeutique , Poumon , Macrophages
6.
Respir Res ; 23(1): 202, 2022 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-35945604

RÉSUMÉ

BACKGROUND: The efficacy and safety of complement inhibition in COVID-19 patients is unclear. METHODS: A multicenter randomized controlled, open-label trial. Hospitalized COVID-19 patients with signs of systemic inflammation and hypoxemia (PaO2/FiO2 below 350 mmHg) were randomized (2:1 ratio) to receive standard of care with or without the C5 inhibitor zilucoplan daily for 14 days, under antibiotic prophylaxis. The primary outcome was improvement in oxygenation at day 6 and 15. RESULTS: 81 patients were randomly assigned to zilucoplan (n = 55) or the control group (n = 26). 78 patients were included in the safety and primary analysis. Most were men (87%) and the median age was 63 years. The mean improvement in PaO2/FiO2 from baseline to day 6 was 56.4 mmHg in the zilucoplan group and 20.6 mmHg in the control group (mean difference + 35.8; 95% confidence interval (CI) - 9.4 to 80.9; p = 0.12), an effect also observed at day 15. Day 28 mortality was 9% in the zilucoplan and 21% in the control group (odds ratio 0.4; 95% CI 0.1 to 1.5). At long-term follow up, the distance walked in a 6-min test was 539.7 m in zilucoplan and 490.6 m in the control group (p = 0.18). Zilucoplan lowered serum C5b-9 (p < 0.001) and interleukin-8 (p = 0.03) concentration compared with control. No relevant safety differences between the zilucoplan and control group were identified. CONCLUSION: Administration of zilucoplan to COVID-19 patients in this proof-of-concept randomized trial was well tolerated under antibiotic prophylaxis. While not reaching statistical significance, indicators of respiratory function (PaO2/FiO2) and clinical outcome (mortality and 6-min walk test) suggest that C5 inhibition might be beneficial, although this requires further research in larger randomized studies.


Sujet(s)
Anti-infectieux , Traitements médicamenteux de la COVID-19 , Complément C5 , Inhibiteurs du complément/effets indésirables , Femelle , Humains , Mâle , Adulte d'âge moyen , Peptides cycliques , SARS-CoV-2 , Résultat thérapeutique
7.
Cytokine ; 157: 155934, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35709568

RÉSUMÉ

Despite global vaccination programs, infections with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) continue to cause severe disease with significant morbidity and mortality. Severe coronavirus disease 2019 (COVID-19) is characterized by an exuberant inflammatory response in the lung leading to acute lung injury and consequent gas exchange problems. Complete insights in this hyperinflammatory response are still lacking. However, a thorough understanding of immunopathogenesis of severe COVID-19 is needed to not only develop personalized targeted therapies, but also to identify biomarkers that predict disease outcome and therapeutic responses. Here we review the current evidence that SARS-CoV-2 activates the inflammasome, which is an intracellular multiprotein complex that leads to the activation and secretion of the interleukin (IL)-1 family cytokines, IL-1ß and IL-18, and to a lytic form of cell death, called pyroptosis. Further we discuss the contribution of inflammasomes and IL-1 family cytokines to the immunopathogenesis of COVID-19 and its clinical implications.


Sujet(s)
COVID-19 , Inflammasomes , Interleukine-1/métabolisme , Cytokines , Humains , Inflammasomes/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Pronostic , SARS-CoV-2
8.
J Allergy Clin Immunol ; 149(5): 1655-1665.e5, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35278495

RÉSUMÉ

BACKGROUND: Chronic rhinosinusitis (CRS) is a heterogeneous disease, with patients having either a high or low type 2 inflammatory endotype. Whereas the type 2-high group is well characterized by IL-5 expression, the type 2-low group, consisting of approximately 20% of CRS with and 50% of CRS without nasal polyp patients, lacks a clear biomarker profile and thus specific therapeutic targets. OBJECTIVE: The aim was to identify underlying molecular pathways of type 2-low CRS, as stratification of patients may allow improvement of personalized treatments. METHODS: Luminex assays were performed to analyze proteins in nasal secretions and tissues of CRS patients. Immunostainings were analyzed for differences in neutrophils, granulocyte-colony stimulating factor (G-CSF), and its receptor in nasal tissue. Neutrophils were isolated from blood of healthy volunteers and stimulated with G-CSF. Effects on apoptosis and neutrophil activity were analyzed with flow cytometry. RESULTS: G-CSF was significantly upregulated in nasal tissue and secretion fluid of type 2-low CRS patients compared to type 2-high patients. In nasal polyp tissue of type 2-low patients, a large infiltration of neutrophils expressing both G-CSF and its receptor was detected, suggesting the presence of a neutrophil-intrinsic autocrine survival mechanism. In response to G-CSF, neutrophils were in an activated state and were resistant to apoptosis, possibly contributing to a chronic inflammation. Of interest, type 2-high nasal polyp patients treated with IgE-blocking omalizumab had increased G-CSF concentrations compared to before treatment. CONCLUSION: G-CSF is an important cytokine regulating neutrophils in type 2-low CRS and has potential in the diagnosis and therapy of the disease.


Sujet(s)
Polypes du nez , Sinusite , Maladie chronique , Facteur de stimulation des colonies de granulocytes , Facteur de stimulation des colonies de granulocytes et de macrophages/métabolisme , Humains , Polypes du nez/traitement médicamenteux , Granulocytes neutrophiles/métabolisme , Sinusite/traitement médicamenteux , Sinusite/métabolisme
10.
J Exp Med ; 219(2)2022 02 07.
Article de Anglais | MEDLINE | ID: mdl-34914824

RÉSUMÉ

In rare instances, pediatric SARS-CoV-2 infection results in a novel immunodysregulation syndrome termed multisystem inflammatory syndrome in children (MIS-C). We compared MIS-C immunopathology with severe COVID-19 in adults. MIS-C does not result in pneumocyte damage but is associated with vascular endotheliitis and gastrointestinal epithelial injury. In MIS-C, the cytokine release syndrome is characterized by IFNγ and not type I interferon. Persistence of patrolling monocytes differentiates MIS-C from severe COVID-19, which is dominated by HLA-DRlo classical monocytes. IFNγ levels correlate with granzyme B production in CD16+ NK cells and TIM3 expression on CD38+/HLA-DR+ T cells. Single-cell TCR profiling reveals a skewed TCRß repertoire enriched for TRBV11-2 and a superantigenic signature in TIM3+/CD38+/HLA-DR+ T cells. Using NicheNet, we confirm IFNγ as a central cytokine in the communication between TIM3+/CD38+/HLA-DR+ T cells, CD16+ NK cells, and patrolling monocytes. Normalization of IFNγ, loss of TIM3, quiescence of CD16+ NK cells, and contraction of patrolling monocytes upon clinical resolution highlight their potential role in MIS-C immunopathogenesis.


Sujet(s)
COVID-19/complications , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Interféron gamma/métabolisme , Cellules tueuses naturelles/immunologie , Monocytes/métabolisme , Récepteurs du fragment Fc des IgG/métabolisme , Syndrome de réponse inflammatoire généralisée/immunologie , Lymphocytes T/immunologie , Adolescent , Pneumocytes/anatomopathologie , Lymphocytes B/immunologie , Vaisseaux sanguins/anatomopathologie , COVID-19/immunologie , COVID-19/anatomopathologie , Prolifération cellulaire , Enfant , Études de cohortes , Activation du complément , Cytokines/métabolisme , Entérocytes/anatomopathologie , Femelle , Humains , Immunité humorale , Inflammation/anatomopathologie , Interféron de type I/métabolisme , Interleukine-15/métabolisme , Activation des lymphocytes/immunologie , Mâle , Récepteurs aux antigènes des cellules T/métabolisme , SARS-CoV-2/immunologie , Superantigènes/métabolisme , Syndrome de réponse inflammatoire généralisée/anatomopathologie
11.
Lancet Respir Med ; 9(12): 1427-1438, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34756178

RÉSUMÉ

BACKGROUND: Infections with SARS-CoV-2 continue to cause significant morbidity and mortality. Interleukin (IL)-1 and IL-6 blockade have been proposed as therapeutic strategies in COVID-19, but study outcomes have been conflicting. We sought to study whether blockade of the IL-6 or IL-1 pathway shortened the time to clinical improvement in patients with COVID-19, hypoxic respiratory failure, and signs of systemic cytokine release syndrome. METHODS: We did a prospective, multicentre, open-label, randomised, controlled trial, in hospitalised patients with COVID-19, hypoxia, and signs of a cytokine release syndrome across 16 hospitals in Belgium. Eligible patients had a proven diagnosis of COVID-19 with symptoms between 6 and 16 days, a ratio of the partial pressure of oxygen to the fraction of inspired oxygen (PaO2:FiO2) of less than 350 mm Hg on room air or less than 280 mm Hg on supplemental oxygen, and signs of a cytokine release syndrome in their serum (either a single ferritin measurement of more than 2000 µg/L and immediately requiring high flow oxygen or mechanical ventilation, or a ferritin concentration of more than 1000 µg/L, which had been increasing over the previous 24 h, or lymphopenia below 800/mL with two of the following criteria: an increasing ferritin concentration of more than 700 µg/L, an increasing lactate dehydrogenase concentration of more than 300 international units per L, an increasing C-reactive protein concentration of more than 70 mg/L, or an increasing D-dimers concentration of more than 1000 ng/mL). The COV-AID trial has a 2 × 2 factorial design to evaluate IL-1 blockade versus no IL-1 blockade and IL-6 blockade versus no IL-6 blockade. Patients were randomly assigned by means of permuted block randomisation with varying block size and stratification by centre. In a first randomisation, patients were assigned to receive subcutaneous anakinra once daily (100 mg) for 28 days or until discharge, or to receive no IL-1 blockade (1:2). In a second randomisation step, patients were allocated to receive a single dose of siltuximab (11 mg/kg) intravenously, or a single dose of tocilizumab (8 mg/kg) intravenously, or to receive no IL-6 blockade (1:1:1). The primary outcome was the time to clinical improvement, defined as time from randomisation to an increase of at least two points on a 6-category ordinal scale or to discharge from hospital alive. The primary and supportive efficacy endpoints were assessed in the intention-to-treat population. Safety was assessed in the safety population. This study is registered online with ClinicalTrials.gov (NCT04330638) and EudraCT (2020-001500-41) and is complete. FINDINGS: Between April 4, and Dec 6, 2020, 342 patients were randomly assigned to IL-1 blockade (n=112) or no IL-1 blockade (n=230) and simultaneously randomly assigned to IL-6 blockade (n=227; 114 for tocilizumab and 113 for siltuximab) or no IL-6 blockade (n=115). Most patients were male (265 [77%] of 342), median age was 65 years (IQR 54-73), and median Systematic Organ Failure Assessment (SOFA) score at randomisation was 3 (2-4). All 342 patients were included in the primary intention-to-treat analysis. The estimated median time to clinical improvement was 12 days (95% CI 10-16) in the IL-1 blockade group versus 12 days (10-15) in the no IL-1 blockade group (hazard ratio [HR] 0·94 [95% CI 0·73-1·21]). For the IL-6 blockade group, the estimated median time to clinical improvement was 11 days (95% CI 10-16) versus 12 days (11-16) in the no IL-6 blockade group (HR 1·00 [0·78-1·29]). 55 patients died during the study, but no evidence for differences in mortality between treatment groups was found. The incidence of serious adverse events and serious infections was similar across study groups. INTERPRETATION: Drugs targeting IL-1 or IL-6 did not shorten the time to clinical improvement in this sample of patients with COVID-19, hypoxic respiratory failure, low SOFA score, and low baseline mortality risk. FUNDING: Belgian Health Care Knowledge Center and VIB Grand Challenges program.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux/usage thérapeutique , Traitements médicamenteux de la COVID-19 , Syndrome de libération de cytokines , Insuffisance respiratoire , Sujet âgé , Belgique , Syndrome de libération de cytokines/traitement médicamenteux , Syndrome de libération de cytokines/virologie , Femelle , Ferritines , Humains , Hypoxie , Interleukine-1/antagonistes et inhibiteurs , Interleukine-6/antagonistes et inhibiteurs , Mâle , Adulte d'âge moyen , Oxygène , Études prospectives , Insuffisance respiratoire/traitement médicamenteux , Insuffisance respiratoire/virologie , SARS-CoV-2 , Résultat thérapeutique
12.
Front Immunol ; 11: 596761, 2020.
Article de Anglais | MEDLINE | ID: mdl-33329586

RÉSUMÉ

The disease course of COVID-19 in patients with immunodeficiencies is unclear, as well as the optimal therapeutic strategy. We report a case of a 37-year old male with common variable immunodeficiency disorder and a severe SARS-CoV-2 infection. After administration of convalescent plasma, the patient's condition improved rapidly. Despite clinical recovery, viral RNA remained detectable up to 60 days after onset of symptoms. We propose that convalescent plasma might be considered as a treatment option in patients with CVID and severe COVID-19. In addition, in patients with immunodeficiencies, a different clinical course is possible, with prolonged viral shedding.


Sujet(s)
Anticorps antiviraux/administration et posologie , COVID-19/thérapie , Déficit immunitaire commun variable , ARN viral , SARS-CoV-2 , Excrétion virale , Adulte , COVID-19/sang , COVID-19/immunologie , Déficit immunitaire commun variable/sang , Déficit immunitaire commun variable/immunologie , Déficit immunitaire commun variable/thérapie , Humains , Immunisation passive , Mâle , ARN viral/sang , ARN viral/immunologie , SARS-CoV-2/immunologie , SARS-CoV-2/métabolisme , Excrétion virale/effets des médicaments et des substances chimiques , Excrétion virale/immunologie , Sérothérapie COVID-19
13.
Trials ; 21(1): 934, 2020 Nov 19.
Article de Anglais | MEDLINE | ID: mdl-33213529

RÉSUMÉ

OBJECTIVES: Zilucoplan (complement C5 inhibitor) has profound effects on inhibiting acute lung injury post COVID-19, and can promote lung repair mechanisms that lead to improvement in lung oxygenation parameters. The purpose of this study is to investigate the efficacy and safety of Zilucoplan in improving oxygenation and short- and long-term outcome of COVID-19 patients with acute hypoxic respiratory failure. TRIAL DESIGN: This is a phase 2 academic, prospective, 2:1 randomized, open-label, multi-center interventional study. PARTICIPANTS: Adult patients (≥18y old) will be recruited at specialized COVID-19 units and ICUs at 9 Belgian hospitals. The main eligibility criteria are as follows: 1) Inclusion criteria: a. Recent (≥6 days and ≤16 days) SARS-CoV-2 infection. b. Chest CT scan showing bilateral infiltrates within the last 2 days prior to randomisation. c. Acute hypoxia (defined as PaO2/FiO2 below 350 mmHg or SpO2 below 93% on minimal 2 L/min supplemental oxygen). d. Signs of cytokine release syndrome characterized by either high serum ferritin, or high D-dimers, or high LDH or deep lymphopenia or a combination of those. 2) Exclusion criteria: e. Mechanical ventilation for more than 24 hours prior to randomisation. f. Active bacterial or fungal infection. g. History of meningococcal disease (due to the known high predisposition to invasive, often recurrent meningococcal infections of individuals deficient in components of the alternative and terminal complement pathways). INTERVENTION AND COMPARATOR: Patients in the experimental arm will receive daily 32,4 mg Zilucoplan subcutaneously and a daily IV infusion of 2g of the antibiotic ceftriaxone for 14 days (or until hospital discharge, whichever comes first) in addition to standard of care. These patients will receive additional prophylactic antibiotics until 14 days after the last Zilucoplan dose: hospitalized patients will receive a daily IV infusion of 2g of ceftriaxone, discharged patients will switch to daily 500 mg of oral ciprofloxacin. The control group will receive standard of care and a daily IV infusion of 2g of ceftriaxone for 1 week (or until hospital discharge, whichever comes first), to control for the effects of antibiotics on the clinical course of COVID-19. MAIN OUTCOMES: The primary endpoint is the improvement of oxygenation as measured by mean and/or median change from pre-treatment (day 1) to post-treatment (day 6 and 15 or at discharge, whichever comes first) in PaO2/FiO2 ratio, P(A-a)O2 gradient and a/A PO2 ratio. (PAO2= Partial alveolar pressure of oxygen, PaO2=partial arterial pressure of oxygen, FiO2=Fraction of inspired oxygen). RANDOMISATION: Patients will be randomized in a 2:1 ratio (Zilucoplan: control). Randomization will be done using an Interactive Web Response System (REDCap). BLINDING (MASKING): In this open-label trial neither participants, caregivers, nor those assessing the outcomes will be blinded to group assignment. NUMBERS TO BE RANDOMISED (SAMPLE SIZE): A total of 81 patients will be enrolled: 54 patients will be randomized to the experimental arm and 27 patients to the control arm. TRIAL STATUS: ZILU-COV protocol Version 4.0 (June 10 2020). Participant recruitment started on June 23 2020 and is ongoing. Given the uncertainty of the pandemic, it is difficult to predict the anticipated end date. TRIAL REGISTRATION: The trial was registered on Clinical Trials.gov on May 11th, 2020 (ClinicalTrials.gov Identifier: NCT04382755 ) and on EudraCT (Identifier: 2020-002130-33 ). FULL PROTOCOL: The full protocol is attached as an additional file, accessible from the Trials website (Additional file 1). In the interest in expediting dissemination of this material, the familiar formatting has been eliminated; this Letter serves as a summary of the key elements of the full protocol.


Sujet(s)
Complément C5/antagonistes et inhibiteurs , Infections à coronavirus/complications , Hypoxie/traitement médicamenteux , Pneumopathie virale/complications , Insuffisance respiratoire/traitement médicamenteux , Maladie aigüe , Adulte , Antibactériens/administration et posologie , Antibactériens/usage thérapeutique , Belgique/épidémiologie , Betacoronavirus/isolement et purification , COVID-19 , Études cas-témoins , Ceftriaxone/administration et posologie , Ceftriaxone/usage thérapeutique , Infections à coronavirus/épidémiologie , Infections à coronavirus/physiopathologie , Infections à coronavirus/virologie , Syndrome de libération de cytokines/sang , Syndrome de libération de cytokines/traitement médicamenteux , Association de médicaments , Humains , Perfusions veineuses , Injections sous-cutanées , Oxygène/sang , Pandémies , Pneumopathie virale/épidémiologie , Pneumopathie virale/physiopathologie , Pneumopathie virale/virologie , Études prospectives , SARS-CoV-2 , Sécurité , Résultat thérapeutique
14.
Trials ; 21(1): 468, 2020 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-32493441

RÉSUMÉ

OBJECTIVES: The purpose of this study is to test the safety and effectiveness of individually or simultaneously blocking IL-6, IL-6 receptor and IL-1 versus standard of care on blood oxygenation and systemic cytokine release syndrome in patients with COVID-19 coronavirus infection and acute hypoxic respiratory failure and systemic cytokine release syndrome. TRIAL DESIGN: A phase 3 prospective, multi-center, interventional, open label, 6-arm 2x2 factorial design study. PARTICIPANTS: Subjects will be recruited at the specialized COVID-19 wards and/or ICUs at 16 Belgian participating hospitals. Only adult (≥18y old) patients will be recruited with recent (≤16 days) COVID-19 infection and acute hypoxia (defined as PaO2/FiO2 below 350mmHg or PaO2/FiO2 below 280 on supplemental oxygen and immediately requiring high flow oxygen device or mechanical ventilation) and signs of systemic cytokine release syndrome characterized by high serum ferritin, or high D-dimers, or high LDH or deep lymphopenia or a combination of those, who have not been on mechanical ventilation for more than 24 hours before randomisation. Patients should have had a chest X-ray and/or CT scan showing bilateral infiltrates within the last 2 days before randomisation. Patients with active bacterial or fungal infection will be excluded. INTERVENTION AND COMPARATOR: Patients will be randomized to 1 of 5 experimental arms versus usual care. The experimental arms consist of Anakinra alone (anti-IL-1 binding the IL-1 receptor), Siltuximab alone (anti-IL-6 chimeric antibody), a combination of Siltuximab and Anakinra, Tocilizumab alone (humanised anti-IL-6 receptor antibody) or a combination of Anakinra with Tocilizumab in addition to standard care. Patients treated with Anakinra will receive a daily subcutaneous injection of 100mg for a maximum of 28 days or until hospital discharge, whichever comes first. Siltuximab (11mg/kg) or Tocilizumab (8mg/kg, with a maximum dose of 800mg) are administered as a single intravenous injection immediately after randomization. MAIN OUTCOMES: The primary end point is the time to clinical improvement defined as the time from randomization to either an improvement of two points on a six-category ordinal scale measured daily till day 28 or discharge from the hospital or death. This ordinal scale is composed of (1) Death; (2) Hospitalized, on invasive mechanical ventilation or ECMO; (3) Hospitalized, on non-invasive ventilation or high flow oxygen devices; (4) Hospitalized, requiring supplemental oxygen; (5) Hospitalized, not requiring supplemental oxygen; (6) Not hospitalized. RANDOMISATION: Patients will be randomized using an Interactive Web Response System (REDCap). A 2x2 factorial design was selected with a 2:1 randomization regarding the IL-1 blockade (Anakinra) and a 1:2 randomization regarding the IL-6 blockade (Siltuximab and Tocilizumab). BLINDING (MASKING): In this open-label trial neither participants, caregivers, nor those assessing the outcomes are blinded to group assignment. NUMBERS TO BE RANDOMISED (SAMPLE SIZE): A total of 342 participants will be enrolled: 76 patients will receive usual care, 76 patients will receive Siltuximab alone, 76 patients will receive Tocilizumab alone, 38 will receive Anakinra alone, 38 patients will receive Anakinra and Siltuximab and 38 patients will receive Anakinra and Tocilizumab. TRIAL STATUS: COV-AID protocol version 3.0 (15 Apr 2020). Participant recruitment is ongoing and started on April 4th 2020. Given the current decline of the COVID-19 pandemic in Belgium, it is difficult to anticipate the rate of participant recruitment. TRIAL REGISTRATION: The trial was registered on Clinical Trials.gov on April 1st, 2020 (ClinicalTrials.gov Identifier: NCT04330638) and on EudraCT on April 3rd 2020 (Identifier: 2020-001500-41). FULL PROTOCOL: The full protocol is attached as an additional file, accessible from the Trials website (Additional file 1). In the interest in expediting dissemination of this material, the familiar formatting has been eliminated; this Letter serves as a summary of the key elements of the full protocol.


Sujet(s)
Anti-inflammatoires/usage thérapeutique , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux/usage thérapeutique , Betacoronavirus/effets des médicaments et des substances chimiques , Antagoniste du récepteur à l'interleukine-1/usage thérapeutique , Anti-inflammatoires/effets indésirables , Anticorps monoclonaux/effets indésirables , Anticorps monoclonaux humanisés/effets indésirables , Belgique , Betacoronavirus/immunologie , Betacoronavirus/pathogénicité , COVID-19 , Essais cliniques de phase III comme sujet , Infections à coronavirus/sang , Infections à coronavirus/diagnostic , Infections à coronavirus/immunologie , Infections à coronavirus/virologie , Association de médicaments , Interactions hôte-pathogène , Humains , Antagoniste du récepteur à l'interleukine-1/effets indésirables , Interleukine-1/antagonistes et inhibiteurs , Interleukine-1/sang , Interleukine-1/immunologie , Interleukine-6/antagonistes et inhibiteurs , Interleukine-6/sang , Interleukine-6/immunologie , Études multicentriques comme sujet , Pandémies , Pneumopathie virale/sang , Pneumopathie virale/diagnostic , Pneumopathie virale/immunologie , Pneumopathie virale/virologie , Études prospectives , Essais contrôlés randomisés comme sujet , Récepteurs à l'interleukine-6/antagonistes et inhibiteurs , Récepteurs à l'interleukine-6/sang , Récepteurs à l'interleukine-6/immunologie , SARS-CoV-2 , Indice de gravité de la maladie , Facteurs temps , Résultat thérapeutique
17.
Trials ; 21(1): 491, 2020 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-32503663

RÉSUMÉ

OBJECTIVES: The hypothesis of the proposed intervention is that Granulocyte-macrophage colony-stimulating factor (GM-CSF) has profound effects on antiviral immunity, and can provide the stimulus to restore immune homeostasis in the lung with acute lung injury post COVID-19, and can promote lung repair mechanisms, that lead to a 25% improvement in lung oxygenation parameters. Sargramostim is a man-made form of the naturally-occurring protein GM-CSF. TRIAL DESIGN: A phase 4 academic, prospective, 2 arm (1:1 ratio), randomized, open-label, controlled trial. PARTICIPANTS: Patients aged 18-80 years admitted to specialized COVID-19 wards in 5 Belgian hospitals with recent (< 2 weeks prior to randomization) confirmed COVID-19 infection and acute respiratory failure defined as a PaO2/FiO2 below 350 mmHg or SpO2 below 93% on minimal 2 L/min supplemental oxygen. Patients were excluded from the trial in case of (1) known serious allergic reactions to yeast-derived products, (2) lithium carbonate therapy, (3) mechanical ventilation prior to randomization, (4) peripheral white blood cell count above 25.000/µL and/or active myeloid malignancy, (5) high dose systemic steroid therapy (> 20 mg methylprednisolone or equivalent), (6) enrolment in another investigational study, (7) pregnant or breastfeeding or (8) ferritin levels > 2000 µg/mL. INTERVENTION AND COMPARATOR: Inhaled sargramostim 125 µg twice daily for 5 days in addition to standard care. Upon progression of disease requiring mechanical ventilation or to acute respiratory distress syndrome (ARDS) and initiation of mechanical ventilator support within the 5 day period, inhaled sargramostim will be replaced by intravenous sargramostim 125 µg/m2 body surface area once daily until the 5 day period is reached. From day 6 onwards, progressive patients in the active group will have the option to receive an additional 5 days of IV sargramostim, based on the treating physician's assessment. Intervention will be compared to standard of care. Subjects progressing to ARDS and requiring invasive mechanical ventilatory support, from day 6 onwards in the standard of care group will have the option (clinician's decision) to initiate IV sargramostim 125m µg/m2 body surface area once daily for 5 days. MAIN OUTCOMES: The primary endpoint of this intervention is measuring oxygenation after 5 days of inhaled (and intravenous) treatment through assessment of a change in pretreatment and post-treatment ratio of PaO2/FiO2 and through measurement of the P(A-a)O2 gradient (PAO2= Partial alveolar pressure of oxygen, PaO2=Partial arterial pressure of oxygen; FiO2= Fraction of inspired oxygen). RANDOMISATION: Patients will be randomized in a 1:1 ratio. Randomization will be done using REDCap (electronic IWRS system). BLINDING (MASKING): In this open-label trial neither participants, caregivers, nor those assessing the outcomes will be blinded to group assignment. NUMBERS TO BE RANDOMISED (SAMPLE SIZE): A total of 80 patients with confirmed COVID-19 and acute hypoxic respiratory failure will be enrolled, 40 in the active and 40 in the control group. TRIAL STATUS: SARPAC protocol Version 2.0 (April 15 2020). Participant recruitment is ongoing in 5 Belgian Hospitals (i.e. University Hospital Ghent, AZ Sint-Jan Bruges, AZ Delta Roeselare, University Hospital Brussels and ZNA Middelheim Antwerp). Participant recruitment started on March 26th 2020. Given the current decline of the COVID-19 pandemic in Belgium, it is difficult to anticipate the rate of participant recruitment. TRIAL REGISTRATION: The trial was registered on Clinical Trials.gov on March 30th, 2020 (ClinicalTrials.gov Identifier: NCT04326920) - retrospectively registered; https://clinicaltrials.gov/ct2/show/NCT04326920?term=sarpac&recrs=ab&draw=2&rank=1 and on EudraCT on March 24th, 2020 (Identifier: 2020-001254-22). FULL PROTOCOL: The full protocol is attached as an additional file, accessible from the Trials website (Additional file 1). In the interest in expediting dissemination of this material, the familiar formatting has been eliminated; this Letter serves as a summary of the key elements of the full protocol.


Sujet(s)
Betacoronavirus , Infections à coronavirus/complications , Facteur de stimulation des colonies de granulocytes et de macrophages/usage thérapeutique , Pneumopathie virale/complications , Essais contrôlés randomisés comme sujet , Insuffisance respiratoire/traitement médicamenteux , Maladie aigüe , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , COVID-19 , Essais cliniques de phase IV comme sujet , Femelle , Humains , Mâle , Adulte d'âge moyen , Oxygène/sang , Pandémies , Études prospectives , Protéines recombinantes/usage thérapeutique , Ventilation artificielle , SARS-CoV-2 , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...