1
|
Shumaker AH, Bhimraj A. Pharmacologic Treatment and Management of Coronavirus Disease 2019. Infect Dis Clin North Am 2025; 39:275-291. [PMID: 40089443 DOI: 10.1016/j.idc.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Widespread vaccination, hybrid immunity, and reduced pathogenicity with circulating Omicron variants have decreased the rate of severe coronavirus disease 2019 (COVID-19) outcomes in the general population. Certain patients with COVID-19 remain at high risk for severe outcomes. Clinicians must individualize treatments based on expected benefits and relative harms for patients with mild-to-moderate COVID-19. Guideline-directed therapy for severe and critical COVID-19 has remained static over the last couple of years. Data on immunomodulatory agents have improved our understanding of the management of severe and critical COVID-19, yet uncertainty remains on the role and timing of these agents in the Omicron era.
Collapse
Affiliation(s)
- Amy Hirsch Shumaker
- Department of Pharmacy, VA Northeast Ohio Healthcare System, OH, USA; School of Medicine, Case Western Reserve University, OH, USA.
| | - Adarsh Bhimraj
- Division of Infectious Diseases, Houston Methodist Hospital, 6560 Fannin Street Scurlock tower, Suite 1512, Houston, TX 77030, USA
| |
Collapse
|
2
|
Belvederi F, Leggeri S, Urbani A, Baroni S. suPAR as a biomarker of support in different clinical settings. Clin Chim Acta 2025; 573:120303. [PMID: 40222544 DOI: 10.1016/j.cca.2025.120303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
The urokinase-type plasminogen activator receptor (uPAR) system, which includes protease, receptor and inhibitors, is essential for key cellular functions like immune activation, cell migration, and tissue remodeling. Soluble uPAR (suPAR), released into circulation, serves as a valuable biomarker for systemic inflammation and immune activation. Elevated suPAR levels are associated with disease severity in conditions such as infections, sepsis, cardiovascular diseases, renal injury, cancer, and autoimmune diseases providing prognostic value especially in acute settings. Recent advancements in diagnostic methods, have enhanced the accuracy of suPAR measurement in serum and plasma. New rapid tests, such as suPARnostic Quick Triage, as well as turbidimetric assays, further expand its clinical applicability. In this review, we discuss the suPAR biomarker, focusing on its biochemical structure, biological functions, measurement methods and areas of clinical interest in different fields of medicine.
Collapse
Affiliation(s)
- Fabio Belvederi
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Simone Leggeri
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy; Unit of Chemistry, Biochemistry and Molecular Biology, "A. Gemelli" Hospital Foundation IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Silvia Baroni
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy; Unit of Chemistry, Biochemistry and Molecular Biology, "A. Gemelli" Hospital Foundation IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy.
| |
Collapse
|
3
|
Mascolo R, Wu MA, Berra S, Vidali M, Pancrazi M, Trotta L, Ceriani E, Negro E, Serati L, Carrozzo L, Calabrò E, Ceriotti F, Andreis A, Collini V, Imazio M, Brucato A. Recurrent pericarditis with normal C-reactive protein: Clinical and laboratory features, biomarkers and outcome in a non-inflammatory phenotype. Eur J Intern Med 2025; 135:108-117. [PMID: 40113467 DOI: 10.1016/j.ejim.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/23/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Differences in recurrent pericarditis with normal vs. elevated C-reactive protein (CRP) are unknown. MATERIALS AND METHODS We studied 448 patients with recurrent or incessant pericarditis. CRP levels <10 mg/L were considered normal. Forty-one patients with normal CRP were tested for interleukin 6 (IL-6), serum amyloid A (SAA) and soluble urokinase plasminogen activator receptor (suPAR). RESULTS Among the cohort (median age 43 years, 48.4% females), 336 patients (75%) had elevated CRP, while 112 (25%) had normal levels. CRP-negative patients were younger (38.6 vs. 43.5 years, p=0.007) and predominantly female (67.9% vs. 42.0%, p<0.001). They presented less often with fever (23.2% vs. 74.1%, p<0.001), pleural involvement (7.1% vs. 61.6% p<0.001), and neutrophilic leucocytosis (WBC 6760 vs. 12315/µL, p<0.001). Pericardiocentesis was performed rarely in CRP-negative patients (2.7% vs. 13.4%, p=0.001). Recurrence rates were similar (5.4 vs. 5.5/100 months-patient, p=0.918). Among CRP-negative patients, 53 (47.3%) had an incessant course characterized by persistent symptoms (pain and tachycardia); abnormal instrumental findings included ECG changes (28.6%), mild pericardial effusion (86.6%), and cardiac magnetic resonance evidence of effusion/oedema/late gadolinium enhancement (43.7%). Anakinra was administered to 48 CRP-positive (14.3%) and 10 CRP-negative patients (8.9%), leading to good responses with discontinuation of NSAIDs and corticosteroids in 39/48 (81.2%) and 5/10 (50.0%), respectively. Among the 41 CRP-negative patients tested, IL-6 and suPAR levels were always normal, while SAA was elevated in 17.1%. CONCLUSIONS Pericarditis with normal CRP exhibits distinct clinical and laboratory features, often presenting with an incessant course. Although rarely elevated, SAA may help to identify inflammation beyond CRP.
Collapse
Affiliation(s)
- Ruggiero Mascolo
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy.
| | - Maddalena Alessandra Wu
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Silvia Berra
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Matteo Vidali
- Clinical Pathology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Massimo Pancrazi
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Lucia Trotta
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Elisa Ceriani
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Enrica Negro
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Lisa Serati
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Luisa Carrozzo
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Elisa Calabrò
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Ferruccio Ceriotti
- Clinical Pathology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Alessandro Andreis
- Department of Cardiology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Valentino Collini
- Cardiothoracic Department, University Hospital Santa Maria della Misericordia, ASUFC, Udine, Italy
| | - Massimo Imazio
- Cardiothoracic Department, University Hospital Santa Maria della Misericordia, ASUFC, Udine, Italy
| | - Antonio Brucato
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
4
|
Nègre P, Tayac D, Jamme T, Combis MS, Maupas-Schwalm F. Early suPAR levels as a predictor of COVID-19 severity: A new tool for efficient patient triage. Infect Dis Now 2025; 55:105058. [PMID: 40101896 DOI: 10.1016/j.idnow.2025.105058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/18/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Following several waves of the COVID-19 pandemic, we are now facing a lower but persistent rate of SARS-CoV-2 infections, with seasonal resurgences often coinciding with other respiratory tract infections. OBJECTIVE We aimed to identify early clinico-biological variables predictive of an unfavorable outcome in patients with primary SARS-CoV-2 infection. We also evaluated the role of suPAR, an innovative biomarker, in predicting disease severity. METHODS We included 255 patients with PCR-confirmed primary SARS-CoV-2 infection and with a 30-day follow-up minimum. Blood samples were collected within the first 24 h of hospitalization to measure suPAR levels. Comprehensive data from medical records were analyzed to assess their predictive value in stratifying patients into seven severity groups, with groups 1 to 3 representing severe COVID-19 (death, intubation, ECMO, or non-invasive ventilation). RESULTS Early plasma suPAR levels were significantly associated with severe disease progression, as evidenced by ANOVA and logistic regression models, highlighting suPAR as a persistent predictive factor for unfavorable outcomes. CONCLUSION Our findings suggest that a single suPAR measurement, performed early after a positive PCR test for SARS-CoV-2, holds strong predictive value for patient outcomes. This biomarker, alongside pulse oximetry and CT scan results, could be instrumental in early patient triage during seasonal COVID-19 resurgences.
Collapse
Affiliation(s)
- Pauline Nègre
- Faculty of Pharmacy, Toulouse III University, France; Medical Biochemistry Laboratory, CHU Toulouse, France
| | - Didier Tayac
- Medical Biochemistry Laboratory, CHU Toulouse, France
| | - Thibaut Jamme
- Medical Biochemistry Laboratory, CHU Toulouse, France
| | | | - Françoise Maupas-Schwalm
- Medical Biochemistry Laboratory, CHU Toulouse, France; Faculty of Medicine, Toulouse III University, France.
| |
Collapse
|
5
|
Patrascu R, Dumitru CS. Advances in Understanding Inflammation and Tissue Damage: Markers of Persistent Sequelae in COVID-19 Patients. J Clin Med 2025; 14:1475. [PMID: 40094929 PMCID: PMC11900244 DOI: 10.3390/jcm14051475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/19/2025] Open
Abstract
This review explores the crucial role of established and emerging biomarkers in the diagnosis, management, and understanding of post-COVID-19 conditions. With COVID-19 affecting multiple organ systems, biomarkers have been instrumental in identifying ongoing inflammation and tissue damage, facilitating early diagnosis and prognostication. Specifically, markers like C-reactive protein (CRP), interleukin-6 (IL-6), and novel entities such as soluble urokinase plasminogen activator receptor (suPAR) and neutrophil extracellular traps (NETs) provide insights into the pathophysiological mechanisms and predict long-term outcomes. This review highlights the integration of these biomarkers into clinical workflows and their implications for personalized medicine, emphasizing their potential in guiding therapeutic interventions and monitoring recovery. Future directions suggest a focus on longitudinal studies to explore biomarker trajectories and their interaction with therapeutic outcomes, aiming to enhance the management of post-COVID-19 conditions and refine public health strategies.
Collapse
Affiliation(s)
- Raul Patrascu
- Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Cristina Stefania Dumitru
- Department of Microscopic Morphology/Histology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| |
Collapse
|
6
|
Wozniak P, Sieminski M, Pyrzowski J, Petrosjan R, Głogowski-Kulasza J, Leszczyński-Czeczatka J. Soluble Urokinase Plasminogen Activator Receptor: A Promising Biomarker for Mortality Prediction Among Critical ED Patients. Int J Mol Sci 2025; 26:1609. [PMID: 40004075 PMCID: PMC11855880 DOI: 10.3390/ijms26041609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Patients admitted to the emergency department (ED) are a highly diverse group in terms of the risk of death. In overcrowded EDs, it becomes crucial to quickly and reliably estimate the risk of death or significant health deterioration. For this purpose, the concentration of soluble urokinase plasminogen activator receptor (suPAR) in plasma has been studied in recent years in various patient populations. In the present study, we tested the hypothesis that measuring suPAR upon the ED admission of critically ill patients can identify those at the highest mortality risk. To verify this hypothesis, we analyzed the relationship between suPAR plasma concentration, other biochemical parameters, and Early Warning Scores (EWSs) on admission and survival to hospital discharge. The study group consisted of 61 ED patients with priority 1 in the Manchester Triage System (MTS), excluding patients with illness caused by environmental factors. Positive correlations between suPAR and inflammatory parameters such as CRP and PCT, as well as the warning scales MEWS, MEDS, and qSOFA, were confirmed. Plasma suPAR concentration on admission was found to be a promising predictor of in-hospital mortality. The study indicated the potential prognostic value of suPAR as the mortality risk predictor for a specific population of critically ill ED patients.
Collapse
Affiliation(s)
| | - Mariusz Sieminski
- Department of Emergency Medicine, Medical University of Gdansk, 80-210 Gdansk, Poland; (P.W.); (J.P.); (R.P.); (J.G.-K.); (J.L.-C.)
| | | | | | | | | |
Collapse
|
7
|
Obeagu EI. The dynamic role of soluble urokinase plasminogen activator receptor (suPAR) in monitoring coagulation dysfunction during COVID-19 progression: a review. Ann Med Surg (Lond) 2025; 87:635-640. [PMID: 40110334 PMCID: PMC11918634 DOI: 10.1097/ms9.0000000000002791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 11/20/2024] [Indexed: 03/22/2025] Open
Abstract
COVID-19, caused by the severe acute respiratory syndrome coronavirus 2, has led to significant morbidity and mortality worldwide, with severe complications often involving coagulation dysfunction and thromboembolic events. Identifying reliable biomarkers for early detection and monitoring of these complications is crucial for improving patient outcomes. The soluble urokinase plasminogen activator receptor (suPAR) has emerged as a potential biomarker in this context, given its role in inflammation and immune response. Elevated suPAR levels correlate with disease severity and inflammatory markers, making it a valuable indicator of the complex interplay between inflammation and coagulation observed in COVID-19. Elevated suPAR levels have been linked to an increased risk of thromboembolic events, such as deep vein thrombosis and pulmonary embolism. Combining suPAR measurement with other coagulation markers, such as D-dimer, enhances the predictive accuracy for thrombotic complications. Furthermore, higher suPAR levels are associated with increased disease severity, intensive care requirements, and higher mortality rates, underscoring its significance in risk stratification and therapeutic decision-making. The integration of suPAR measurement into routine clinical practice for COVID-19 could significantly aid in early diagnosis, risk assessment, and monitoring of therapeutic interventions. By providing insights into the patient's inflammatory and coagulation status, suPAR can guide the timely initiation of anticoagulant therapy and other treatments aimed at reducing thromboembolic complications. As research continues to validate suPAR's utility across diverse populations and clinical settings, it holds promise for becoming an integral component of clinical management strategies to mitigate the morbidity and mortality associated with COVID-19.
Collapse
|
8
|
Viermyr HK, Tonby K, Ponzi E, Trouillet-Assant S, Poissy J, Arribas JR, Dyon-Tafani V, Bouscambert-Duchamp M, Assoumou L, Halvorsen B, Tekin NB, Diallo A, De Gastines L, Munthe LA, Murphy SL, Ueland T, Michelsen AE, Lund-Johansen F, Aukrust P, Mootien J, Dervieux B, Zerbib Y, Richard JC, Prével R, Malvy D, Timsit JF, Peiffer-Smadja N, Roux D, Piroth L, Ait-Oufella H, Vieira C, Dalgard O, Heggelund L, Müller KE, Møller JH, Kildal AB, Skogen V, Aballi S, Sjøberg Øgaard JD, Dyrhol-Riise AM, Tveita A, Alirezaylavasani A, Costagliola D, Yazdanpanah Y, Olsen IC, Dahl TB, Kared H, Holten AR, Trøseid M. Safety of baricitinib in vaccinated patients with severe and critical COVID-19 sub study of the randomised Bari-SolidAct trial. EBioMedicine 2025; 111:105511. [PMID: 39731852 PMCID: PMC11743795 DOI: 10.1016/j.ebiom.2024.105511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND The Bari-SolidAct randomized controlled trial compared baricitinib with placebo in patients with severe COVID-19. A post hoc analysis revealed a higher incidence of serious adverse events (SAEs) among SARS-CoV-2-vaccinated participants who had received baricitinib. This sub-study aimed to investigate whether vaccination influences the safety profile of baricitinib in patients with severe COVID-19. METHODS Biobanked samples from 146 participants (55 vaccinated vs. 91 unvaccinated) were analysed longitudinally for inflammation markers, humoral responses, tissue viral loads, and plasma viral antigens on days 1, 3, and 8. High-dimensional analyses, including RNA sequencing and flow cytometry, were performed on available samples. Mediation analyses were used to assess relationships between SAEs, baseline-adjusted biomarkers, and treatment-vaccination status. FINDINGS Vaccinated participants were older, more frequently hospitalized, had more comorbidities, and exhibited higher nasopharyngeal viral loads. Baricitinib treatment did not affect antibody responses or viral clearance, but reduced markers of T-cell and monocyte activation compared to placebo (sCD25, sCD14, sCD163, sTIM-3). Age, baseline levels of plasma viral antigen, and several inflammatory markers, as well as IL-2, IL-6, Neopterin, CXCL16, sCD14, and suPAR on day 8 were associated with the occurrence of SAEs. However, mediation analyses of markers linked to SAEs, baricitinib treatment, or vaccination status did not reveal statistically significant interactions between vaccination status and SAEs. INTERPRETATION This sub-study did not identify any virus- or host-related biomarkers significantly associated with the interaction between SARS-CoV-2 vaccination status and the safety of baricitinib. However, caution should be exercised due to the moderate sample size. FUNDING EU Horizon 2020 (grant number 101015736).
Collapse
Affiliation(s)
- Hans-Kittil Viermyr
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section for Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kristian Tonby
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Erica Ponzi
- Department of Research Support for Clinical Trials, Oslo University Hospital, Oslo, Norway
| | - Sophie Trouillet-Assant
- Centre International de Recherche en Infectiologie (CIRI), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France; Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| | - Julien Poissy
- Université Lille, Inserm U1285, CHU Lille, Pôle de Médecine Intensive-Réanimatin, CNRS, UMR 8576, France; Université Lille, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), F-59000, Lille, France
| | - José R Arribas
- Infectious Diseases Unit, Internal Medicine Department, La Paz University Hospital, IdiPAZ, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Virginie Dyon-Tafani
- Centre International de Recherche en Infectiologie (CIRI), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
| | - Maude Bouscambert-Duchamp
- Hospices Civils de Lyon, Laboratoire de Virologie, Institut des Agents Infectieux de Lyon, Centre National de Référence des Virus Respiratoires France Sud, F-69317, Lyon, France; Université Claude Bernard Lyon 1, Virpath, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, F-69372, Lyon, France
| | - Lambert Assoumou
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique (IPLESP), Paris, France
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nuriye Basdag Tekin
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alpha Diallo
- ANRS, Maladies Infectieuses Emergentes, F-75015, Paris, France; Institut National de la Santé et de la Recherche Médicale, INSERM, F-75013, Paris, France
| | - Lucie De Gastines
- ANRS, Maladies Infectieuses Emergentes, F-75015, Paris, France; Institut National de la Santé et de la Recherche Médicale, INSERM, F-75013, Paris, France
| | - Ludvig A Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sarah Louise Murphy
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Joy Mootien
- Intensive Care Unit, Antibiotic Stewardship Team, Groupe Hospitalier Région Mulhouse et Sud Alsace, Mulhouse, France
| | - Benjamin Dervieux
- Infectious Diseases Unit, Groupe Hospitalier Région Mulhouse et Sud Alsace, Mulhouse, France
| | - Yoann Zerbib
- Intensive Care Department, Amiens-Picardie University Hospital, Amiens, France
| | - Jean-Christophe Richard
- Medical Intensive Care Unit, Hospices Civils de Lyon, Croix-Rousse Hospital - Université Lyon 1, Lyon, France; CREATIS INSERM 1044 CNRS 5220, Villeurbanne, France
| | - Renaud Prével
- CHU Bordeaux, Medical Intensive Care Unit, Bordeaux, France; Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France
| | - Denis Malvy
- Department of Infectious and Tropical Diseases, University Hospital, UMR 1219 Inserm/EMR 271 IRD, University of Bordeaux, Bordeaux, France
| | - Jean-François Timsit
- AP-HP, Bichat Hospital, Medical and Infectious Diseases ICU (MI2), F-75018, Paris, France; Université Paris-Cité, INSERM, F-75018, Paris, France; OUCTOME REA Research Network, France
| | - Nathan Peiffer-Smadja
- Université Paris Cité, Inserm, IAME, Paris, 75018, France; Service de Maladies Infectieuses et Tropicales, Hôpital Bichat Claude Bernard, AP-HP, Paris, 75018, France
| | - Damien Roux
- Université Paris Cité, AP-HP, Hôpital Louis Mourier, DMU ESPRIT, Service de Médecine Intensive Réanimation, Colombes, France
| | - Lionel Piroth
- Infectious Diseases Department, University Hospital, Dijon, France; INSERM CIC 1432, University of Burgundy, Dijon, France
| | - Hafid Ait-Oufella
- Service de Médecine Intensive-Réanimation, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, F-75012, France
| | - Cesar Vieira
- Centro Hospitalar Universitário de Lisboa Central, Hospital Curry Cabral, Department of Intensive Care Medicine - Lisbon, Portugal
| | - Olav Dalgard
- Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway; Department of Infectious Diseases, Akershus University Hospital, Norway
| | - Lars Heggelund
- Department of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Norway; Department of Clinical Science, Bergen Integrated Diagnostic Stewardship Cluster, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Karl Erik Müller
- Department of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Norway; Department of Clinical Science, Bergen Integrated Diagnostic Stewardship Cluster, Faculty of Medicine, University of Bergen, Bergen, Norway
| | | | - Anders Benjamin Kildal
- Department of Anesthesiology and Intensive Care, University Hospital of North Norway, Tromsø, Norway; Faculty of Health Sciences, UIT The Arctic University of Norway, Norway
| | - Vegard Skogen
- Department of Infectious Diseases, University Hospital of North Norway, Tromsø, Norway; Faculty of Health Sciences, UIT The Arctic University of Norway, Norway
| | - Saad Aballi
- Department of Infectious Diseases, Østfold Hospital Kalnes, Grålum, Norway
| | - Jonas Daniel Sjøberg Øgaard
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Anders Tveita
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section for Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway; Department of Internal Medicine, Bærum Hospital, Vestre Viken Hospital Trust, 1346, Gjettum, Norway
| | | | - Dominique Costagliola
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique (IPLESP), Paris, France
| | - Yazdan Yazdanpanah
- Infectious and Tropical Diseases Department, Bichat - Claude Bernard Hospital, AP-HP Nord-Université Paris Cité, Paris, France; IAME INSERM UMR 1137, Université Paris Cité, Paris, France; ANRS, Maladies Infectieuses Emergentes, F-75015, Paris, France; Institut National de la Santé et de la Recherche Médicale, INSERM, F-75013, Paris, France
| | - Inge Christoffer Olsen
- Department of Research Support for Clinical Trials, Oslo University Hospital, Oslo, Norway
| | - Tuva Børresdatter Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Aleksander Rygh Holten
- Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway; Department of Acute Medicine, Oslo University Hospital, Oslo, Norway
| | - Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section for Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway.
| |
Collapse
|
9
|
Kyriazopoulou E, Akinosoglou K, Florou E, Kouriannidi E, Bogosian A, Tsachouridou O, Syrigos KN, Gatselis N, Milionis H, Papanikolaou IC, Sympardi S, Dafni M, Alevizou A, Amvrazi AV, Alexandrou E, Archontoulis K, Argyraki K, Alexiou Z, Georgiou Y, Gkogka D, Kyrailidi F, Kalyva V, Nikolopoulou T, Ioannou S, Bakakos P, Karathanassiou G, Koklanos K, Miletis DN, Tili AM, Vakkas L, Vila I, Panagopoulos P, Samarkos M, Chrysos G, Dalekos GN, Poulakou G, Metallidis S, Giamarellos-Bourboulis EJ. Anakinra efficacy in COVID-19 pneumonia guided by soluble urokinase plasminogen activator receptor: Association with the inflammatory burden of the host. Int J Antimicrob Agents 2025; 65:107405. [PMID: 39647797 DOI: 10.1016/j.ijantimicag.2024.107405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Anakinra was approved by the European Medicines Agency and received Emergency Use Authorization by the United States Food and Drug Administration for patients with COVID-19 pneumonia at risk for severe respiratory failure (SRF) with blood levels of soluble urokinase plasminogen activator receptor (suPAR) ≥ 6 ng/mL. We report the final results of the phase II open-label single-arm SAVE trial in a large population. METHODS Patients with COVID-19 pneumonia and suPAR levels ≥ 6 ng/mL received subcutaneous anakinra 100 mg once daily for 10 days. The primary outcome was the incidence of SRF by day 14. Secondary outcomes were 30-day mortality, incidence of SRF according to time delay for start of treatment, safety, and associations with the inflammatory burden of the host. RESULTS From March 2020 to March 2022, a total of 992 patients were enrolled. The incidence of SRF was 18.8%, similar to the results of the phase III pivotal SAVE-MOREtrial. The overall 30-day mortality was 9.5%. Participants were divided into 4 subgroups according to time delay between symptoms onset and start of anakinra. The incidence of SRF was similar for all subgroups. Serious adverse events were reported in 15.4%; only 3 were possibly related to anakinra. The most common adverse event was increased liver function tests. A post hoc comparison with the pivotal phase III trial showed similar anakinra outcomes among patient subgroups by levels of inflammatory mediators and D-dimers. CONCLUSIONS Results support the efficacy of anakinra as being similar to that of the pivotal registrational trial for COVID-19 pneumonia. The lack of a comparator group is a limitation. TRIAL REGISTRATION ClinicalTrials.gov, NCT04357366.
Collapse
Affiliation(s)
- Evdoxia Kyriazopoulou
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - Eleni Florou
- Hellenic Institute for the Study of Sepsis, Athens, Greece
| | - Elli Kouriannidi
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - Olga Tsachouridou
- First Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Konstantinos N Syrigos
- Third Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Nikolaos Gatselis
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, Full Member of the European Reference Network on Hepatological Diseases (ERN RARE-LIVER), General University Hospital of Larissa, Larissa, Greece
| | - Haralampos Milionis
- First Department of Internal Medicine, University of Ioannina, Medical School, Ioannina, Greece
| | - Ilias C Papanikolaou
- Department of Pulmonary Medicine, General Hospital of Corfu "Agia Eirini", Greece
| | - Styliani Sympardi
- First Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Maria Dafni
- First Department of Internal Medicine, Korgialeneion-Benakeion General Hospital, Athens, Greece
| | - Antonia Alevizou
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Alexia-Vasiliki Amvrazi
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Errika Alexandrou
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Kyprianos Archontoulis
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Katerina Argyraki
- Department of Internal Medicine, Sotiria Athens Hospital of Chest Diseases, Athens, Greece
| | - Zoi Alexiou
- Second Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Yakinthi Georgiou
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Dimitra Gkogka
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Foteini Kyrailidi
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Vassiliki Kalyva
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - Sofia Ioannou
- Department of Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Petros Bakakos
- First Department of Pulmonary Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Karathanassiou
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Kyriakos Koklanos
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Dionysios-Nikolaos Miletis
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Anna-Maria Tili
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Lampros Vakkas
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Ioanna Vila
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Periklis Panagopoulos
- Second Department of Internal Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Michael Samarkos
- First Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - George Chrysos
- Second Department of Internal Medicine, Tzaneio General Hospital of Piraeus, Athens, Greece
| | - George N Dalekos
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, Full Member of the European Reference Network on Hepatological Diseases (ERN RARE-LIVER), General University Hospital of Larissa, Larissa, Greece
| | - Garyphallia Poulakou
- Third Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Symeon Metallidis
- First Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelos J Giamarellos-Bourboulis
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece; Hellenic Institute for the Study of Sepsis, Athens, Greece.
| |
Collapse
|
10
|
Li F, Tian X, Wang L, Wu LP, Liu X, Peng HY. Role of plasma suPAR, sTNFR1, and sTNFR2 levels in risk stratification and outcome prediction of complicated acute kidney injury in elderly patients with coronavirus disease 2019. Exp Gerontol 2024; 198:112634. [PMID: 39561952 DOI: 10.1016/j.exger.2024.112634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
OBJECTIVE The aim of this study is to investigate the early prognostic efficacy of plasma soluble urokinase-type plasminogen activator receptor (suPAR), soluble tumor necrosis factor receptor 1 (sTNFR1), and soluble tumor necrosis factor receptor 2 (sTNFR2) in complicated acute kidney injury (AKI) in patients with coronavirus disease 2019 (COVID-19), and to analyze the relevant factors contributing to complicated AKI in these patients. METHODS Patients with COVID-19 hospitalized at the Affiliated Baiyun Hospital of Guizhou Medical University from March 2022 to March 2024 were selected as study participants. A total of 589 patients met the inclusion and exclusion criteria, 68 patients complicated with AKI were classified as AKI group, and the remaining 521 cases were divided into proportion sampling method and randomly selected 200 samples, which were classified as non-AKI group. Additionally, 50 healthy controls were enrolled as the control group. Logistic regression analysis was conducted to identify the relevant factors associated with complicated AKI in patients with COVID-19. Receiver operating characteristic (ROC) curves were plotted to evaluate the prognostic efficacy of plasma suPAR, sTNFR1, and sTNFR2 indicators for complicated AKI in patients with COVID-19. RESULTS Among the patients with COVID-19 in the AKI group, 43 were males (63.20 %), with a median age of 79.00 (interquartile range: 75.00, 83.00) years, while the non-AKI group comprised 83 males (41.50 %), with a median age of 73.00 (interquartile range: 60.00, 80.75) years. Comparison of the sex and age between the two groups indicated that males and elderly patients had increased risks of complicated AKI (P < 0.05). Plasma levels of suPAR, sTNFR1, and sTNFR2 in the AKI group were significantly higher than those in the non-AKI group (P < 0.05). Logistic regression analysis indicated that suPAR and sTNFR2 were independent factors influencing complicated AKI in patients with COVID-19 (P < 0.05). The ROC curve for a single indicator showed that suPAR had the highest prognostic efficacy for complicated AKI, with an area under the curve (AUC) of 0.813, a sensitivity of 79.4 %, and a specificity of 74.0 %. The combined use of suPAR and sTNFR2 for risk assessment yielded the highest AUC of 0.838, with a sensitivity of 66.2 % and a specificity of 87.5 %. The combined risk assessment using all three indicators (suPAR, sTNFR1, and sTNFR2) had an AUC of 0.837, with a sensitivity of 64.7 % and a specificity of 89.0 %. CONCLUSION Elderly patients had increased risks of complicated AKI. Indicators such as suPAR, sTNFR1, and sTNFR2 can assist in assessing the risk in patients with COVID-19 complicated AKI, with suPAR demonstrating the highest prognostic efficacy as a single indicator. The combined detection of suPAR, sTNFR1, and sTNFR2 offers greater prognostic value than using any single indicator.
Collapse
Affiliation(s)
- Fang Li
- Demartment of Nerhrology, The Affiliated Hospital of Guizhou Medical University, Guizhou 550004, China
| | - Xue Tian
- Demartment of Nerhrology, The Affiliated Hospital of Guizhou Medical University, Guizhou 550004, China
| | - Lu Wang
- Department of Nephrology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Ling-Pei Wu
- Department of Nephrology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Xiao Liu
- Department of Nephrology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Hong-Ying Peng
- Department of Nephrology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China.
| |
Collapse
|
11
|
Sehgal T, Anand A, Vijayabupathy G, Khan M. Soluble Urokinase Plasminogen Activator Receptor: A Useful Marker for Predicting Mortality in COVID-19 Patients. Cureus 2024; 16:e74438. [PMID: 39723295 PMCID: PMC11669473 DOI: 10.7759/cureus.74438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction Efficient and practical healthcare based on prognostic indicators can reduce morbidity and mortality in hospitalized COVID-19 patients. Soluble urokinase plasminogen activator receptor (suPAR) predicts clinical outcomes and respiratory failure in SARS-CoV-2 patients, but additional research is needed. Among other characteristics, we aimed to evaluate the predictive value of suPAR in COVID-19 patients. Methods This observational study was conducted at the All India Institute of Medical Sciences in New Delhi between January and April 2022. Patients within the age range from 18 to 85 years with mild, moderate, or severe COVID-19 infections were included in the study. Twenty-one patients (group 1) had positive RT-PCR throat and nasal swabs. Nine patients (group 2) with fever but without COVID-19 were recruited as controls. Ninety patient samples were tested for suPAR on days 1, 5, and 10 utilizing suPARnostic AUTO Flex ELISA kits (ViroGates, Denmark). Results The median age was 59 years in both groups. COVID-19 was mild in six patients (29%), moderate in seven patients (33%), and severe in eight patients (38%). At a median follow-up of 10 days, 8 out of 21 patients (38%) in group 1 died, while none in group 2 died. Patients' median suPAR levels were 4.35 ng/ml on day 1, 4.68 ng/ml on day 5, and 4.37 ng/ml on day 10. In the control group, suPAR levels were 1.6 ng/ml on day 1, 1.7 ng/ml on day 5, and 2.02 ng/ml on day 10. The suPAR levels were statistically significant on day 1 (p = 0.004), day 5 (p = 0.04), and day 10 (p = 0.007) between the patient and the control groups. Conclusion Patients who died had higher suPAR levels on days 1 (p = 0.007), 5 (p = 0.01), and 10 (p = 0.03) than survivors. The suPAR cut-off ≥ 3.64 (AUC = 0.82) predicts mortality with 88% sensitivity and 73% specificity.
Collapse
Affiliation(s)
- Tushar Sehgal
- Laboratory Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Ambika Anand
- Laboratory Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Gowtham Vijayabupathy
- Laboratory Medicine, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Maroof Khan
- Biostatistics, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| |
Collapse
|
12
|
Arientová S, Matúšková K, Bartoš O, Beran O, Holub M. Evaluation of Soluble Urokinase Plasminogen Activator Receptor in COVID-19 Patients. J Clin Med 2024; 13:6340. [PMID: 39518479 PMCID: PMC11546495 DOI: 10.3390/jcm13216340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/04/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: This retrospective study analyzed soluble urokinase plasminogen activator receptor (suPAR) plasma levels alongside routine inflammatory markers, including the neutrophil-to-lymphocyte count ratio, C-reactive protein (CRP), interleukin-6 (IL-6), procalcitonin (PCT), and D-dimers in COVID-19 patients hospitalized during the Omicron wave of the pandemic. Methods: We measured plasma suPAR levels using a suPARnostic® Quick Triage kit. We divided COVID-19 patients into two groups based on the severity of SARS-CoV-2 infection according to the National Institutes of Health (NIH) criteria. The logistic regression analysis tested the predictive value of the biomarkers. Results: We evaluated 160 consecutive COVID-19 patients hospitalized between January and August 2022. The cohort exhibited a high incidence of comorbidities, with an in-hospital mortality rate of 5.6%. Upon admission, the median suPAR plasma levels were not significantly different between patients with mild COVID-19 (n = 110) and those with moderate/severe disease (n = 50), with 7.25 ng/mL and 7.55 ng/mL, respectively. We observed significant differences (p < 0.01) between the groups for CRP and IL-6 levels that were higher in moderate/severe disease than in mild infection. Additionally, suPAR plasma levels were above the normal range (0-2.00 ng/mL) in all patients, with a significant positive correlation identified between suPAR levels and serum IL-6, PCT, and creatinine levels. Conclusions: These findings indicate that COVID-19 during the Omicron wave is strongly associated with elevated suPAR levels; however, these levels do not directly correlate with the severity of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Simona Arientová
- Department of Infectious Diseases, Military University Hospital Prague and First Faculty of Medicine of Charles University, 16902 Prague, Czech Republic; (S.A.); (K.M.); (O.B.)
| | - Kateřina Matúšková
- Department of Infectious Diseases, Military University Hospital Prague and First Faculty of Medicine of Charles University, 16902 Prague, Czech Republic; (S.A.); (K.M.); (O.B.)
| | - Oldřich Bartoš
- Military Health Institute, Military Medical Agency, 16200 Prague, Czech Republic
| | - Ondřej Beran
- Department of Infectious Diseases, Military University Hospital Prague and First Faculty of Medicine of Charles University, 16902 Prague, Czech Republic; (S.A.); (K.M.); (O.B.)
| | - Michal Holub
- Department of Infectious Diseases, Military University Hospital Prague and First Faculty of Medicine of Charles University, 16902 Prague, Czech Republic; (S.A.); (K.M.); (O.B.)
- Ústřední Vojenská Nemocnice—Vojenská Fakultní Nemonice Praha, U Vojenské Nemocnice 1200, Praha 6-Břevnov, 16902 Prague, Czech Republic
| |
Collapse
|
13
|
Weckman AM, Guagliardo SAJ, Crowley VM, Moro L, Piubelli C, Ursini T, van Ierssel SH, Gobbi FG, Emetulu H, Rizwan A, Angelo KM, Licitra C, Connor BA, Barkati S, Ngai M, Zhong K, Huits R, Hamer DH, Libman M, Kain KC. Host Response Markers of Inflammation and Endothelial Activation Associated with COVID-19 Severity and Mortality: A GeoSentinel Prospective Observational Cohort. Viruses 2024; 16:1615. [PMID: 39459948 PMCID: PMC11512287 DOI: 10.3390/v16101615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND The effect of the COVID-19 pandemic on healthcare systems emphasized the need for rapid and effective triage tools to identify patients at risk of severe or fatal infection. Measuring host response markers of inflammation and endothelial activation at clinical presentation may help to inform appropriate triage and care practices in patients with SARS-CoV-2 infection. METHODS We enrolled patients with COVID-19 across five GeoSentinel clinical sites (in Italy, Belgium, Canada, and the United States) from September 2020 to December 2021, and analyzed the association of plasma markers, including soluble urokinase-type plasminogen activator receptor (suPAR), soluble tumor necrosis factor receptor-1 (sTREM-1), interleukin-6 (IL-6), interleukin-8 (IL-8), complement component C5a (C5a), von Willebrand factor (VWF-a2), and interleukin-1 receptor antagonist (IL-1Ra), with 28-day (D28) mortality and 7-day (D7) severity (discharged, hospitalized on ward, or died/admitted to the ICU). RESULTS Of 193 patients, 8.9% (16 of 180) died by D28. Higher concentrations of suPAR were associated with increased odds of mortality at D28 and severity at D7 in univariable and multivariable regression models. The biomarkers sTREM-1 and IL-1Ra showed bivariate associations with mortality at D28 and severity at D7. IL-6, VWF, C5a, and IL-8 were not as indicative of progression to severe disease or death. Conclusions: Our findings confirm previous studies' assertions that point-of-care tests for suPAR and sTREM-1 could facilitate the triage of patients with SARS-CoV-2 infection, which may help guide hospital resource allocation.
Collapse
Affiliation(s)
- Andrea M. Weckman
- UHN-Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada; (A.M.W.)
| | | | - Valerie M. Crowley
- UHN-Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada; (A.M.W.)
| | - Lucia Moro
- Department of Infectious Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
| | - Chiara Piubelli
- Department of Infectious Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
| | - Tamara Ursini
- Department of Infectious Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
| | - Sabrina H. van Ierssel
- Department of Internal Medicine, Antwerp University Hospital (UZA), 2650 Antwerp, Belgium
| | - Federico G. Gobbi
- Department of Infectious Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
| | - Hannah Emetulu
- International Society of Travel Medicine, Atlanta, GA 30338, USA
| | - Aisha Rizwan
- International Society of Travel Medicine, Atlanta, GA 30338, USA
| | - Kristina M. Angelo
- Division of Global Migration and Quarantine, Travelers’ Health Branch, Atlanta, GA 30322, USA
| | - Carmelo Licitra
- Orlando Health Travel Medicine and Infectious Disease, Orlando, FL 34761, USA
| | - Bradley A. Connor
- Weill Cornell Medical College and the New York Center for Travel and Tropical Medicine, New York, NY 10022, USA
| | - Sapha Barkati
- J.D. MacLean Centre for Tropical Diseases, McGill University, Montreal, QC H3A 0G4, Canada
| | - Michelle Ngai
- UHN-Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada; (A.M.W.)
| | - Kathleen Zhong
- UHN-Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada; (A.M.W.)
| | - Ralph Huits
- Department of Infectious Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
- Institute of Tropical Medicine Antwerp, 2000 Antwerp, Belgium
| | - Davidson H. Hamer
- Section of Infectious Diseases, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Center on Emerging Infectious Diseases, Boston University, Boston, MA 02118, USA
- Department of Global Health, Boston University School of Public Health, Boston, MA 02118, USA
| | - Michael Libman
- J.D. MacLean Centre for Tropical Diseases, McGill University, Montreal, QC H3A 0G4, Canada
| | - Kevin C. Kain
- UHN-Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada; (A.M.W.)
- Division of Infectious Diseases, Department of Medicine, MaRS Centre, TMDT, University of Toronto, 10th Floor 10-351, Toronto, QC M5G 1L7, Canada
| |
Collapse
|
14
|
Alkhaleq HA, Hacker I, Karram T, Hamoud S, Kabala A, Abassi Z. Potential Nephroprotective Effect of uPA against Ischemia/Reperfusion-Induced Acute Kidney Injury in αMUPA Mice and HEK-293 Cells. Biomedicines 2024; 12:2323. [PMID: 39457635 PMCID: PMC11505258 DOI: 10.3390/biomedicines12102323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: The incidence of acute kidney injury (AKI) has been steadily increasing. Despite its high prevalence, there is no pathogenetically rational therapy for AKI. This deficiency stems from the poor understanding of the pathogenesis of AKI. Renal ischemia/hypoxia is one of the leading causes of clinical AKI. This study investigates whether αMUPA mice, overexpressing the urokinase plasminogen activator (uPA) gene are protected against ischemic AKI, thus unraveling a potential renal damage treatment target. Methods: We utilized an in vivo model of I/R-induced AKI in αMUPA mice and in vitro experiments of uPA-treated HEK-293 cells. We evaluated renal injury markers, histological changes, mRNA expression of inflammatory, apoptotic, and autophagy markers, as compared with wild-type animals. Results: the αMUPA mice exhibited less renal injury post-AKI, as was evident by lower SCr, BUN, and renal NGAL and KIM-1 along attenuated adverse histological alterations. Notably, the αMUPA mice exhibited decreased levels pro-inflammatory, fibrotic, apoptotic, and autophagy markers like TGF-β, IL-6, STAT3, IKB, MAPK, Caspase-3, and LC3. By contrast, ACE-2, p-eNOS, and PGC1α were higher in the kidneys of the αMUPA mice. In vitro results of the uPA-treated HEK-293 cells mirrored the in vivo findings. Conclusions: These results indicate that uPA modulates key pathways involved in AKI, offering potential therapeutic targets for mitigating renal damage.
Collapse
Affiliation(s)
- Heba Abd Alkhaleq
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel; (H.A.A.); (I.H.); (A.K.)
| | - Israel Hacker
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel; (H.A.A.); (I.H.); (A.K.)
| | - Tony Karram
- Department of Vascular Surgery, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Shadi Hamoud
- Internal Medicine E, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Aviva Kabala
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel; (H.A.A.); (I.H.); (A.K.)
| | - Zaid Abassi
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel; (H.A.A.); (I.H.); (A.K.)
- Laboratory Medicine, Rambam Health Care Campus, Haifa 3109601, Israel
| |
Collapse
|
15
|
Christaki M, Samanidou V, Liontos A, Konstantopoulou R, Milionis H. Post-COVID-19 Multisystem Inflammatory Syndrome in Adults (MIS-A) With Elevated Levels of Soluble Urokinase Plasminogen Activator Receptor (suPAR) Treated With Anakinra: A Case Report. Cureus 2024; 16:e70848. [PMID: 39493146 PMCID: PMC11531791 DOI: 10.7759/cureus.70848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2024] [Indexed: 11/05/2024] Open
Abstract
The COVID-19 pandemic has brought attention to a newly identified syndrome of multisystem inflammation. This potentially fatal complication of the disease was initially observed in children and later in adults. It affects primarily unvaccinated patients and may manifest within a timeframe of 2-12 weeks following infection. Soluble urokinase plasminogen activator receptor (suPAR), a novel biomarker, highlights the severity of inflammation and the degree of immune system activation. Herein, we report a case of a patient with multisystem inflammatory syndrome in adults (MIS-A) and markedly elevated suPAR levels, successfully treated with interleukin-1 (IL-1) receptor antagonist. A 59-year-old female was admitted to our hospital due to febrile illness (up to 40°C) with chills, vomiting, non-bloody diarrhea, and abdominal pain for four days prior to her admission. She tested positive for SARS-CoV-2 12 weeks before her presentation. During hospitalization, the patient deteriorated clinically with multiorgan involvement and hemodynamically instability, with concomitant markedly elevated inflammatory markers. Extensive workup with high suPAR levels led to post-COVID-19 MIS-A diagnosis, and treatment with dexamethasone and an interleukin-1 receptor antagonist (IL-1ra), anakinra, was administered. The subcutaneous injection of anakinra effectively and safely deterred MIS-A. Further research is needed to investigate the role of interleukin-1 inhibitors for the management of this potentially life-threatening condition.
Collapse
Affiliation(s)
- Maria Christaki
- 1st Division of Internal Medicine and Infectious Diseases Unit, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, GRC
| | - Valentini Samanidou
- 1st Division of Internal Medicine and Infectious Diseases Unit, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, GRC
| | - Angelos Liontos
- 1st Division of Internal Medicine and Infectious Diseases Unit, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, GRC
| | - Revekka Konstantopoulou
- 1st Division of Internal Medicine and Infectious Diseases Unit, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, GRC
| | - Haralampos Milionis
- 1st Division of Internal Medicine and Infectious Diseases Unit, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, GRC
| |
Collapse
|
16
|
Adami ME, Giamarellos-Bourboulis EJ, Polyzogopoulou E. Towards improved point-of-care (POC) testing for patients with suspected sepsis: POC tests for host biomarkers and possible microbial pathogens. Expert Rev Mol Diagn 2024; 24:829-839. [PMID: 39135402 DOI: 10.1080/14737159.2024.2392283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/11/2024] [Indexed: 08/15/2024]
Abstract
INTRODUCTION Sepsis is a heterogeneous syndrome often misdiagnosed. Point-of-care (POC) diagnostic tests are commonly used to guide decision and include host biomarkers and molecular diagnostics. AREAS COVERED The diagnostic and prognostic accuracy of established and emerging biomarkers for sepsis, including procalcitonin (PCT) soluble urokinase plasminogen activator receptor (suPAR), presepsin, TRAIL/IP-10/CRP, MxA, and MxA-CRP, are analyzed in this review. The clinical utility of the two prevalent molecular techniques for pathogens identification using polymerase chain reaction (PCR) assays is also presented: FILMARRAY and QIAstat-Dx RP. EXPERT OPINION The rising benefits of the combined use of POC biomarkers with molecular diagnostics in daily clinical routine appear to outperform conventional practices in terms of reduced turnaround time, timely diagnosis, and prompt administration of the appropriate treatment. Yet, this must be further demonstrated in future investigations. However, the cost-effectiveness of POC tests and the high rate of false positive and negative results, indicate the need for a comprehensive clinical evaluation.
Collapse
Affiliation(s)
- Maria-Evangelia Adami
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - Effie Polyzogopoulou
- Department of Emergency Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| |
Collapse
|
17
|
Louka M, Tatsi EB, Vassiliu S, Theoharis G, Straka K, Filippatos F, Dourdouna MM, Siahanidou T, Syriopoulou V, Michos A. The Soluble Urokinase Plasminogen Activator Receptor as a Severity Biomarker in Children With Acute COVID-19 or Multisystem Inflammatory Syndrome. Pediatr Infect Dis J 2024; 43:477-482. [PMID: 38251905 DOI: 10.1097/inf.0000000000004244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
BACKGROUND Elevated soluble urokinase plasminogen activator receptor (suPAR) has been associated with a poor prognosis in serious infections. The aim of this study was to evaluate the clinical value of suPAR in children with acute coronavirus disease 2019 (COVID-19) or multisystem inflammatory syndrome (MIS-C). METHODS Serum suPAR was measured using the suPARnostic AUTO Flex enzyme-linked immunosorbent assay in hospitalized children with COVID-19, MIS-C, bacterial pneumonia, and healthy controls. RESULTS A total of 211 children with a mean (±SD) age of 6.9 ± 4.96 years were tested; with COVID-19: 59 (28%), MIS-C: 36 (17%), pneumonia: 78 (37%) and healthy controls: 38 (18%). In the acute phase, the levels of suPAR (mean ± SD) were: MIS-C: 8.11 ± 2.80 ng/mL, COVID-19: 4.91 ± 1.90 ng/mL, pneumonia: 4.25 ± 1.44 ng/mL and controls: 2.09 ± 0.47 ng/mL ( P < 0.001). Children with acute COVID-19 and a severe or moderate clinical presentation had higher values than those with mild symptoms: 5.79 ± 1.58 versus 5.40 ± 1.94 versus 3.19 ± 0.73 ng/mL, respectively ( P < 0.001). In the MIS-C group, children hospitalized in the intensive care unit and in need of mechanical ventilation had higher suPAR than those who were not admitted to an intensive care unit: 9.32 ± 3.06 versus 7.13 ± 2.19 ng/mL, respectively ( P = 0.023). In children with COVID-19 or MIS-C, a correlation was detected between suPAR values and length of hospitalization ( rs = 0.418, P < 0.001). CONCLUSIONS The findings suggest that suPAR may be a valuable biomarker of disease severity in children with COVID-19 or MIS-C. This could facilitate the identification of children in need of intensive anti-inflammatory treatment, as it has been shown in adults with severe COVID-19.
Collapse
Affiliation(s)
- Magdalini Louka
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| | - Elizabeth Barbara Tatsi
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
- University Research Institute of Maternal and Child Health and Precision Medicine, Athens, Greece
| | - Sofia Vassiliu
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| | - George Theoharis
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| | - Kalliopi Straka
- Pediatric Intensive Care Unit, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Filippos Filippatos
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| | - Maria Myrto Dourdouna
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| | - Tania Siahanidou
- Neonatal Unit, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Vasiliki Syriopoulou
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| | - Athanasios Michos
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| |
Collapse
|
18
|
Kyriazopoulou E, Dalekos GN, Metallidis S, Poulakou G, Papanikolaou IC, Tzavara V, Argyraki K, Alexiou Z, Panagopoulos P, Samarkos M, Chrysos G, Tseliou A, Milionis H, Sympardi S, Vasishta A, Giamarellos-Bourboulis EJ. HEPARIN-BINDING PROTEIN LEVELS PREDICT UNFAVORABLE OUTCOME IN COVID-19 PNEUMONIA: A POST HOC ANALYSIS OF THE SAVE TRIAL. Shock 2024; 61:395-399. [PMID: 38517242 DOI: 10.1097/shk.0000000000002315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
ABSTRACT We aimed to evaluate heparin-binding protein (HBP) as a marker of prognosis of unfavorable outcome in COVID-19 pneumonia. This was a post hoc analysis of the SAVE clinical trial investigating anakinra treatment, guided by suPAR (soluble urokinase plasminogen activator receptor) levels ≥6 ng/mL, for the prevention of severe respiratory failure in hospitalized patients with COVID-19 pneumonia. Baseline HBP plasma levels were measured in 534 patients by fluorescence dry quantitative immunoassay using the Jet-iStar 800 analyzer. Concentrations higher than 35 ng/mL predicted 30-day mortality with a moderate specificity of 53.3% and negative predictive value 78.1%; sensitivity was low (29.0%). After multivariate Cox analysis, HBP higher than 35 ng/mL was an independent predictor of 30-day unfavorable outcome (adjusted hazard ratio, 1.77; 95% CI, 1.06-2.94; P = 0.028) and these patients were also at greater risk of death after 90 days (hazard ratio, 1.85; 95% CI, 1.25-2.74; P = 0.002). The cutoff was not predictive of development of severe respiratory failure, septic shock or acute kidney injury. Among patients with baseline HBP levels higher than 35 ng/mL, anakinra treatment was associated with decreased mortality (7.2%) versus comparators (18.1%; P < 0.001). Results confirm that HBP may be an early biomarker of poor outcome among preselected patients at risk from COVID-19 pneumonia.ClinicalTrials.gov registration NCT04357366.
Collapse
Affiliation(s)
- Evdoxia Kyriazopoulou
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - George N Dalekos
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, Full Member of the European Reference Network on Hepatological Diseases (ERN RARE-LIVER), General University Hospital of Larissa, Larissa, Greece
| | - Symeon Metallidis
- First Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Garyphalia Poulakou
- Third Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Ilias C Papanikolaou
- Department of Pulmonary Medicine, General Hospital of Corfu "Agia Eirini," Athens, Greece
| | - Vasiliki Tzavara
- First Department of Internal Medicine, Korgialeneion-Benakeion General Hospital, Athens, Greece
| | - Katerina Argyraki
- Department of Internal Medicine, Sotiria Athens Hospital of Chest Diseases, Athens, Greece
| | - Zoi Alexiou
- Second Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Periklis Panagopoulos
- Second Department of Internal Medicine, Democritus University of Thrace, Alexandropoilis, Greece
| | - Michael Samarkos
- First Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - George Chrysos
- Second Department of Internal Medicine, Tzaneio General Hospital of Piraeus, Athens, Greece
| | - Aikaterini Tseliou
- First Department of Internal Medicine, G. Gennimatas General Hospital of Athens, Athens, Greece
| | - Haralampos Milionis
- First Department of Internal Medicine, University of Ioannina, Medical School, Ioannina, Greece
| | - Styliani Sympardi
- First Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | | | | |
Collapse
|
19
|
Marin MJ, van Wijk XMR, Chambliss AB. Advances in sepsis biomarkers. Adv Clin Chem 2024; 119:117-166. [PMID: 38514209 DOI: 10.1016/bs.acc.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Sepsis, a dysregulated host immune response to an infectious agent, significantly increases morbidity and mortality for hospitalized patients worldwide. This chapter reviews (1) the basic principles of infectious diseases, pathophysiology and current definition of sepsis, (2) established sepsis biomarkers such lactate, procalcitonin and C-reactive protein, (3) novel, newly regulatory-cleared/approved biomarkers, such as assays that evaluate white blood cell properties and immune response molecules, and (4) emerging biomarkers and biomarker panels to highlight future directions and opportunities in the diagnosis and management of sepsis.
Collapse
Affiliation(s)
- Maximo J Marin
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | | | - Allison B Chambliss
- Department of Pathology & Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
20
|
Adami ME, Kotsaki A, Antonakos N, Giannitsioti E, Chalvatzis S, Saridaki M, Avgoustou C, Akinosoglou K, Dakou K, Damoraki G, Katrini K, Koufargyris P, Lekakis V, Panagaki A, Safarika A, Eugen-Olsen J, Giamarellos-Bourboulis EJ. qSOFA combined with suPAR for early risk detection and guidance of antibiotic treatment in the emergency department: a randomized controlled trial. Crit Care 2024; 28:42. [PMID: 38321472 PMCID: PMC10848347 DOI: 10.1186/s13054-024-04825-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/01/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Sepsis guidelines suggest immediate start of resuscitation for patients with quick Sequential Organ Failure Assessment (qSOFA) 2 or 3. However, the interpretation of qSOFA 1 remains controversial. We investigated whether measurements of soluble urokinase plasminogen activator receptor (suPAR) may improve risk detection when qSOFA is 1. METHODS The study had two parts. At the first part, the combination of suPAR with qSOFA was analyzed in a prospective cohort for early risk detection. At the second part, the double-blind, randomized controlled trial (RCT) SUPERIOR evaluated the efficacy of the suPAR-guided medical intervention. SUPERIOR took place between November 2018 and December 2020. Multivariate stepwise Cox regression was used for the prospective cohort, while univariate and multivariate logistic regression was used for the RCT. Consecutive admissions at the emergency department (ED) with suspected infection, qSOFA 1 and suPAR ≥ 12 ng/mL were allocated to single infusion of placebo or meropenem. The primary endpoint was early deterioration, defined as at least one-point increase of admission Sequential Organ Failure Assessment (SOFA) score the first 24 h. RESULTS Most of the mortality risk was for patients with qSOFA 2 and 3. Taking the hazard ratio (HR) for death of patients with qSOFA = 1 and suPAR < 12 ng/mL as reference, the HR of qSOFA = 1 and suPAR ≥ 12 ng/mL for 28-day mortality was 2.98 (95% CI 2.11-3.96). The prospective RCT was prematurely ended due to pandemia-related ED re-allocations, with 91 patients enrolled: 47 in the placebo and 44 in the meropenem arm. The primary endpoint was met in 40.4% (n = 19) and 15.9% (n = 7), respectively (difference 24.5% [5.9-40.8]; odds ratio 0.14 [0.04-0.50]). One post hoc analysis showed significant median changes of SOFA score after 72 and 96 h equal to 0 and - 1, respectively. CONCLUSIONS Combining qSOFA 1 with the biomarker suPAR improves its prognostic performance for unfavorable outcome and can help decision for earlier treatment. Trial registration EU Clinical Trials Register (EudraCT, 2018-001008-13) and Clinical-Trials.gov (NCT03717350). Registered 24 October 2018.
Collapse
Affiliation(s)
- Maria Evangelia Adami
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Antigone Kotsaki
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Nikolaos Antonakos
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Efthymia Giannitsioti
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Stamatios Chalvatzis
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Maria Saridaki
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Christina Avgoustou
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | | | | | - Georgia Damoraki
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Konstantina Katrini
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Panagiotis Koufargyris
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Vasileios Lekakis
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Antonia Panagaki
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Asimina Safarika
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Evangelos J Giamarellos-Bourboulis
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece.
- Hellenic Institute for the Study of Sepsis, Athens, Greece.
| |
Collapse
|
21
|
Gallo A, Massaro MG, Camilli S, Di Francesco S, Gerardino L, Verrecchia E, Sicignano LL, Landi F, Manna R, Montalto M. Interleukin-1 Blockers in Recurrent and Acute Pericarditis: State of the Art and Future Directions. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:241. [PMID: 38399529 PMCID: PMC10890577 DOI: 10.3390/medicina60020241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024]
Abstract
Diseases of the pericardium encompass a spectrum of conditions, including acute and recurrent pericarditis, where inflammation plays a pivotal role in the pathogenesis and clinical manifestations. Anti-inflammatory therapy indeed forms the cornerstone of treating these conditions: NSAIDs, colchicine, and corticosteroids (as a second-line treatment) are recommended by current guidelines. However, these medications come with several contraindications and are not devoid of adverse effects. In recent years, there has been an increased focus on the role of the inflammasome and potential therapeutic targets. Recurrent pericarditis also shares numerous characteristics with other autoinflammatory diseases, in which interleukin-1 antagonists have already been employed with good efficacy and safety. The objective of this review is to summarize the available studies on the use of anti-IL-1 drugs both in acute and recurrent pericarditis.
Collapse
Affiliation(s)
- Antonella Gallo
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (L.G.); (E.V.); (L.L.S.); (F.L.); (R.M.); (M.M.)
| | - Maria Grazia Massaro
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Sara Camilli
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Silvino Di Francesco
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Laura Gerardino
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (L.G.); (E.V.); (L.L.S.); (F.L.); (R.M.); (M.M.)
| | - Elena Verrecchia
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (L.G.); (E.V.); (L.L.S.); (F.L.); (R.M.); (M.M.)
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Ludovico Luca Sicignano
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (L.G.); (E.V.); (L.L.S.); (F.L.); (R.M.); (M.M.)
| | - Francesco Landi
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (L.G.); (E.V.); (L.L.S.); (F.L.); (R.M.); (M.M.)
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Raffaele Manna
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (L.G.); (E.V.); (L.L.S.); (F.L.); (R.M.); (M.M.)
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Massimo Montalto
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (L.G.); (E.V.); (L.L.S.); (F.L.); (R.M.); (M.M.)
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
22
|
Vasbinder A, Padalia K, Pizzo I, Machado K, Catalan T, Presswalla F, Anderson E, Ismail A, Hutten C, Huang Y, Blakely P, Azam TU, Berlin H, Feroze R, Launius C, Meloche C, Michaud E, O'Hayer P, Pan M, Shadid HR, Rasmussen LJH, Roberts DA, Zhao L, Banerjee M, Murthy V, Loosen SH, Chalkias A, Tacke F, Reiser J, Giamarellos-Bourboulis EJ, Eugen-Olsen J, Pop-Busui R, Hayek SS. SuPAR, biomarkers of inflammation, and severe outcomes in patients hospitalized for COVID-19: The International Study of Inflammation in COVID-19. J Med Virol 2024; 96:e29389. [PMID: 38235904 PMCID: PMC10829525 DOI: 10.1002/jmv.29389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/13/2023] [Accepted: 12/28/2023] [Indexed: 01/19/2024]
Abstract
Severe coronavirus disease 2019 (COVID-19) is a hyperinflammatory syndrome. The biomarkers of inflammation best suited to triage patients with COVID-19 are unknown. We conducted a prospective multicenter observational study of adult patients hospitalized specifically for COVID-19 from February 1, 2020 to October 19, 2022. Biomarkers measured included soluble urokinase plasminogen activator receptor (suPAR), C-reactive protein, interleukin-6, procalcitonin, ferritin, and D-dimer. In-hospital outcomes examined include death and the need for mechanical ventilation. Patients admitted in the United States (US, n = 1962) were used to compute area under the curves (AUCs) and identify biomarker cutoffs. The combined European cohorts (n = 1137) were used to validate the biomarker cutoffs. In the US cohort, 356 patients met the composite outcome of death (n = 197) or need for mechanical ventilation (n = 290). SuPAR was the most important predictor of the composite outcome and had the highest AUC (0.712) followed by CRP (0.642), ferritin (0.619), IL-6 (0.614), D-dimer (0.606), and lastly procalcitonin (0.596). Inclusion of other biomarkers did not improve discrimination. A suPAR cutoff of 4.0 ng/mL demonstrated a sensitivity of 95.4% (95% CI: 92.4%-98.0%) and negative predictive value (NPV) of 92.5% (95% CI: 87.5%-96.9%) for the composite outcome. Patients with suPAR < 4.0 ng/mL comprised 10.6% of the cohort and had a 0.8% probability of the composite outcome. Applying this cutoff to the validation cohort yielded a sensitivity of 93.8% (90.4%-96.7%) and NPV of 95.5% (93.1%-97.8%) for the composite outcome. Among commonly measured biomarkers, suPAR offered stronger discriminatory ability and may be useful in triaging low-risk patients with COVID-19.
Collapse
Affiliation(s)
- Alexi Vasbinder
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kishan Padalia
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ian Pizzo
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kristen Machado
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Tonimarie Catalan
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Feriel Presswalla
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Elizabeth Anderson
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anis Ismail
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christina Hutten
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yiyuan Huang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Pennelope Blakely
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Tariq U Azam
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hanna Berlin
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Rafey Feroze
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Launius
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Chelsea Meloche
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Erinleigh Michaud
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrick O'Hayer
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael Pan
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Husam R Shadid
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Donald A Roberts
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lili Zhao
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Mousumi Banerjee
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Venkatesh Murthy
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sven H Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Athanasios Chalkias
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Outcomes Research Consortium, Cleveland, Ohio, USA
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | | | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Rodica Pop-Busui
- Division of Endocrinology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
23
|
Helena C, Giulia T, Alessandra C, Roberto G, Caterina S, Sabrina G, Simone B, Tommaso B, Alessandra B, Roberto L. Detection of Inflammatory Biomarker suPAR in COVID-19 Disease With CHORUS TRIO Instrument. Biomark Insights 2023; 18:11772719231210407. [PMID: 38035182 PMCID: PMC10687937 DOI: 10.1177/11772719231210407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 10/11/2023] [Indexed: 12/02/2023] Open
Abstract
Background Deregulation in the urokinase-type plasminogen activator receptor (uPA/uPAR) system is reported in many diseases where the immune system is activated. During SARS-CoV-2 infection, a rise in soluble uPAR (suPAR) levels has been detected and its concentration above 6 µg/L predicts worsening to severe respiratory failure 14 days earlier, with a positive predictive value of 85.9%, and was the prerequisite for a treatment with anakinra, a recombinant IL-1 receptor antagonist that blocks the activity of both IL-1α and IL-1β. Objectives To compare suPAR concentrations measured by CHORUS suPAR on CHORUS TRIO instrument of DIESSE with the commercially available suPARnostic (ViroGates) ELISA assay. Design A single-centre, non-pharmacological, diagnostic study was performed. Results A total of 522 serum samples from patients with COVID-19 were tested for suPAR. CHORUS suPAR resulted accurate and reliable, with a high grade of specificity (97.9%), accuracy (97.3%) and sensitivity (96.9%). The median concentration of suPAR, as determined with CHORUS suPAR, was 6.8 µg/L (interquartile range 4.5-9.7) in patients with moderate disease (n = 465) and 8.5 µg/L (interquartile range 5.4-10.6) in patients with severe disease. Among patients with moderate and severe disease, 60.6% and 71.9%, respectively, reached the cut-off concentration of suPAR ⩾6 µg/L, defining their illness severity and suggesting eligibility to anakinra treatment. Conclusion CHORUS suPAR kit resulted as sensitive, specific, accurate and able to quantify suPAR concentrations in patients with moderate and severe COVID-19.
Collapse
Affiliation(s)
- Cerutti Helena
- R&D DIESSE Diagnostica Senese S.p.A. Società Benefit, Monteriggioni, Siena, Italy
| | - Tesi Giulia
- R&D DIESSE Diagnostica Senese S.p.A. Società Benefit, Monteriggioni, Siena, Italy
| | | | - Guerranti Roberto
- Laboratorio Patologia Clinica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Silvestrini Caterina
- Laboratorio Patologia Clinica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Gori Sabrina
- Laboratorio Patologia Clinica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Bianciardi Simone
- R&D DIESSE Diagnostica Senese S.p.A. Società Benefit, Monteriggioni, Siena, Italy
| | - Bandini Tommaso
- R&D DIESSE Diagnostica Senese S.p.A. Società Benefit, Monteriggioni, Siena, Italy
| | - Brogi Alessandra
- R&D DIESSE Diagnostica Senese S.p.A. Società Benefit, Monteriggioni, Siena, Italy
| | - Leoncini Roberto
- Laboratorio Patologia Clinica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| |
Collapse
|
24
|
Azawi N, Mosholt KSS, Fryd ND, Lund L, Brignone JI, Hvid N, Wulf-Johansson H, Pedersen OBV, Saekmose SG, Dabestani S. suPARnostic: an advanced predictive tool for detecting recurrence in renal cell carcinoma. BMC Urol 2023; 23:168. [PMID: 37875832 PMCID: PMC10594785 DOI: 10.1186/s12894-023-01337-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Plasma soluble urokinase-type Plasminogen Activator Receptor (suPAR) predicts disease aggressiveness in renal cell carcinoma (ccRCC), but its prognostic accuracy has not been investigated. To investigate the prognostic accuracy of preoperative plasma suPAR in patients who received curative treatment for initially localized ccRCC. METHODS We retrospectively analyzed plasma samples stored in the Danish National Biobank between 2010 and 2015 from 235 patients with ccRCC at any stage. Relationships with outcome analyzed using univariate and multiple logistic Cox regression analysis. RESULTS There were 235 patients with ccRCC. The median follow-up period was 7.7 years. In univariate analysis suPAR ≥ 6 ng/mL was significantly associated with overall survival (OS) and recurrence-free survival (RFS). Patients with elevated suPAR were more likely to recur, with a Hazard Ratio (HR) of 2.3 for RFS. In multiple logistic regression, suPAR ≥ 6 ng/mL remained a negative predictor of OS and RFS. Limitations include retrospective study design, wide confidence intervals, and tumor subtype heterogeneity bias. CONCLUSIONS ccRCC patients with high plasma suPAR concentrations are at an elevated risk of disease recurrence and see lower OS. suPAR is a promising surveillance tool to more precisely follow up with ccRCC patients and detect future recurrences. In this study, we showed that new type of liquid marker in blood plasma, called suPAR, is associated to a higher risk of kidney cancer recurrence when elevated above 6ng/mL. We also showed suPAR to independently be able to predict patients overall and recurrence free survival in patient with any stage of kidney cancer.
Collapse
Affiliation(s)
- Nessn Azawi
- Department of Urology, Zealand University Hospital, Sygehusvej 6, Roskilde, 4000, Denmark.
- Institute of Clinical Medicine, University of Copenhagen, Norregade 10, copenhagen, Copenhagen, 1165, Denmark.
| | | | - Nathalie Demuth Fryd
- Deparment of Urology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Aarhus, Denmark
| | - Lars Lund
- Deparment of Urology, Odense University Hospital, J. B. Winslows Vej 4, Odense, 5000, Denmark
| | - Juan Ignacio Brignone
- Department of Urology, Aalborg University hospital, Hobrovej 18-22, Aalborg, Aalborg, 9100, Denmark
| | - Nanna Hvid
- Deparment of Urology, Odense University Hospital, J. B. Winslows Vej 4, Odense, 5000, Denmark
| | - Helle Wulf-Johansson
- Department of Clinical Immunology, Naestved Hospital, Ringstedgade 61, Naestved, Naestved, 4700, Denmark
| | | | - Susanne Gjørup Saekmose
- Department of Clinical Immunology, Naestved Hospital, Ringstedgade 61, Naestved, Naestved, 4700, Denmark
| | - Saeed Dabestani
- Department of Urology, Kristianstad Central Hospital, Region Skane, Kristianstad, Sweden
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Lund, Sweden
| |
Collapse
|
25
|
Zhu K, Mukherjee K, Wei C, Hayek SS, Collins A, Gu C, Corapi K, Altintas MM, Wang Y, Waikar SS, Bianco AC, Koch A, Tacke F, Reiser J, Sever S. The D2D3 form of uPAR acts as an immunotoxin and may cause diabetes and kidney disease. Sci Transl Med 2023; 15:eabq6492. [PMID: 37729431 DOI: 10.1126/scitranslmed.abq6492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is a risk factor for kidney diseases. In addition to suPAR, proteolysis of membrane-bound uPAR results in circulating D1 and D2D3 proteins. We showed that when exposed to a high-fat diet, transgenic mice expressing D2D3 protein developed progressive kidney disease marked by microalbuminuria, elevated serum creatinine, and glomerular hypertrophy. D2D3 transgenic mice also exhibited insulin-dependent diabetes mellitus evidenced by decreased levels of insulin and C-peptide, impaired glucose-stimulated insulin secretion, decreased pancreatic β cell mass, and high fasting blood glucose. Injection of anti-uPAR antibody restored β cell mass and function in D2D3 transgenic mice. At the cellular level, the D2D3 protein impaired β cell proliferation and inhibited the bioenergetics of β cells, leading to dysregulated cytoskeletal dynamics and subsequent impairment in the maturation and trafficking of insulin granules. D2D3 protein was predominantly detected in the sera of patients with nephropathy and insulin-dependent diabetes mellitus. These sera inhibited glucose-stimulated insulin release from human islets in a D2D3-dependent manner. Our study showed that D2D3 injures the kidney and pancreas and suggests that targeting this protein could provide a therapy for kidney diseases and insulin-dependent diabetes mellitus.
Collapse
Affiliation(s)
- Ke Zhu
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Kamalika Mukherjee
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Agnieszka Collins
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Changkyu Gu
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Kristin Corapi
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Yong Wang
- Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
| | - Sushrut S Waikar
- Section of Nephrology, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA 02129, USA
| | - Antonio C Bianco
- Division of Endocrinology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Alexander Koch
- Department of Gastroenterology, Metabolic Diseases and Internal Intensive Care Medicine, University Hospital Aachen, 52072 Aachen, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sanja Sever
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| |
Collapse
|
26
|
Chandna A, Mahajan R, Gautam P, Mwandigha L, Dittrich S, Kumar V, Osborn J, Kumar P, Koshiaris C, Varghese GM, Lubell Y, Burza S. Point-of-care prognostication in moderate Covid-19: Analytical validation and prognostic accuracy of a soluble urokinase plasminogen activator receptor (suPAR) rapid test. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001538. [PMID: 37603548 PMCID: PMC10441780 DOI: 10.1371/journal.pgph.0001538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 07/20/2023] [Indexed: 08/23/2023]
Abstract
The soluble urokinase plasminogen activator receptor (suPAR) has been proposed as a biomarker for risk stratification of patients presenting with acute infections. However, most studies evaluating suPAR have used platform-based assays, the accuracy of which may differ from point-of-care tests capable of informing timely triage in settings without established laboratory capacity. Using samples and data collected during a prospective cohort study of 425 patients presenting with moderate Covid-19 to two hospitals in India, we evaluated the analytical performance and prognostic accuracy of a commercially-available rapid diagnostic test (RDT) for suPAR, using an enzyme-linked immunosorbent assay (ELISA) as the reference standard. Our hypothesis was that the suPAR RDT might be useful for triage of patients presenting with moderate Covid-19 irrespective of its analytical performance when compared with the reference test. Although agreement between the two tests was limited (bias = -2.46 ng/mL [95% CI = -2.65 to -2.27 ng/mL]), prognostic accuracy to predict supplemental oxygen requirement was comparable, whether suPAR was used alone (area under the receiver operating characteristic curve [AUC] of RDT = 0.73 [95% CI = 0.68 to 0.79] vs. AUC of ELISA = 0.70 [95% CI = 0.63 to 0.76]; p = 0.12) or as part of a published multivariable prediction model (AUC of RDT-based model = 0.74 [95% CI = 0.66 to 0.83] vs. AUC of ELISA-based model = 0.72 [95% CI = 0.64 to 0.81]; p = 0.78). Lack of agreement between the RDT and ELISA in our cohort warrants further investigation and highlights the importance of assessing candidate point-of-care tests to ensure management algorithms reflect the assay that will ultimately be used to inform patient care. Availability of a quantitative point-of-care test for suPAR opens the door to suPAR-guided risk stratification of patients with Covid-19 and other acute infections in settings with limited laboratory capacity.
Collapse
Affiliation(s)
- Arjun Chandna
- Cambodia Oxford Medical Research Unit, Angkor Hospital for Children, Siem Reap, Cambodia
- Centre for Tropical Medicine & Global Health, University of Oxford, Oxford, United Kingdom
| | | | - Priyanka Gautam
- Department of Infectious Diseases, Christian Medical College, Vellore, India
| | - Lazaro Mwandigha
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Sabine Dittrich
- Centre for Tropical Medicine & Global Health, University of Oxford, Oxford, United Kingdom
- Foundation for Innovative Diagnostics, Geneva, Switzerland
- Deggendorf Institut of Technology, European-Campus Rottal Inn, Pfarrkirchen, Germany
| | | | | | - Pragya Kumar
- Department of Community & Family Medicine, All India Institute of Medical Sciences, Patna, India
| | - Constantinos Koshiaris
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - George M. Varghese
- Department of Infectious Diseases, Christian Medical College, Vellore, India
| | - Yoel Lubell
- Centre for Tropical Medicine & Global Health, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Sakib Burza
- Médecins Sans Frontières, New Delhi, India
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
27
|
Sixt T, Moretto F, Esteve C, Duong M, Buisson M, Mahy S, Blot M, Piroth L. Healing Treatments in COVID-19 Patients: A Narrative Review. J Clin Med 2023; 12:4672. [PMID: 37510786 PMCID: PMC10380607 DOI: 10.3390/jcm12144672] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Since December 2019, many drugs have been evaluated or advocated as potential treatments of SARS-CoV-2 induced disease (COVID-19), including many repositioned drugs and some others specifically developed for these diseases. They can be roughly classified into three categories according to their main mechanism of action (passive immunization, direct antivirals, and anti-inflammatory treatments), and their use depends on the stage of the disease. Despite often promising preclinical data, most of the treatments evaluated failed to show a significant clinical benefit. In addition, a few others have seen their effectiveness affected by the occurrence of SARS-CoV-2 variants and sub-variants. Herein, the aim of this article is to take stock of the data available as of the 14th of July 2022, concerning the specific healing options evaluated for patients suffering from COVID-19. We focus particularly on healing treatments of COVID-19 and do not deal with preventive treatments such as vaccine. Associated therapies such as venous thromboembolism prophylaxis are not detailed since they are covered in a specific chapter of this issue. Passive immunization, especially through monoclonal antibodies, showed a positive impact on the clinical evolution, whether in outpatients or inpatients without oxygen supply. However, their effectiveness strongly depends on the type of SARS-CoV-2 variant, and often decreases or even vanishes with the most recent variants. Among direct antiviral treatments, ritonavir-boosted nirmatrelvir appears to currently be the cornerstone in the management of early infections, but its use may be limited by drug interactions. Remdesivir remains as an alternative in this situation, even though it is potentially less convenient. Anti-inflammatory treatments have often been shown to be the most effective in inpatients with oxygen supply. Dexamethasone is now a cornerstone of management of these patients. Added tocilizumab seems beneficial in the case of hyper inflammation. JAK inhibitors and anakinra have also gained an interest in some studies. As a conclusion of this narrative review, the best treatment strategy has yet to be defined and is likely to evolve in the future, not only because many other drugs are still under development and evaluation, but also because of the viral epidemics and epidemiology evolution.
Collapse
Affiliation(s)
- Thibault Sixt
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
| | - Florian Moretto
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
| | - Clementine Esteve
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
| | - Michel Duong
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
| | - Marielle Buisson
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
| | - Sophie Mahy
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
| | - Mathieu Blot
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
- CHU Dijon-Bourgogne, INSERM, Université de Bourgogne, CIC 1432, Module Épidémiologie Clinique, 21000 Dijon, France
- Lipness Team, INSERM Research Centre LNC-UMR1231 and LabEx LipSTIC, University of Burgundy, 21078 Dijon, France
| | - Lionel Piroth
- Infectious Diseases Department, Dijon-Bourgogne University Hospital, 21000 Dijon, France
- CHU Dijon-Bourgogne, INSERM, Université de Bourgogne, CIC 1432, Module Épidémiologie Clinique, 21000 Dijon, France
| |
Collapse
|
28
|
Kyriazopoulou E, Giamarellos-Bourboulis EJ, Akinosoglou K. Biomarkers to guide immunomodulatory treatment: where do we stand? Expert Rev Mol Diagn 2023; 23:945-958. [PMID: 37691280 DOI: 10.1080/14737159.2023.2258063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/20/2023] [Accepted: 09/08/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION This review summarizes current progress in the development of biomarkers to guide immunotherapy in oncology, rheumatology, and critical illness. AREAS COVERED An extensive literature search was performed about biomarkers classifying patients' immune responses to guide immunotherapy in oncology, rheumatology, and critical illness. Surface markers, such as programmed death-ligand 1 (PD-L1), genetic biomarkers, such as tumor mutation load, and circulating tumor DNA are biomarkers associated with the effectiveness of immunotherapy in oncology. Genomics, metabolomics, and proteomics play a crucial role in selecting the most suitable therapeutic options for rheumatologic patients. Phenotypes and endotypes are a promising approach to detect critically ill patients with hyper- or hypo-inflammation. Sepsis trials using biomarkers such as ferritin, lymphopenia, HLA-DR expression on monocytes and PD-L1 to guide immunotherapy have been already conducted or are currently ongoing. Immunotherapy in COVID-19 pneumonia, guided by C-reactive protein and soluble urokinase plasminogen activator receptor (suPAR) has improved patient outcomes globally. More research is needed into immunotherapy in other critical conditions. EXPERT OPINION Targeted immunotherapy has improved outcomes in oncology and rheumatology, paving the way for precision medicine in the critically ill. Transcriptomics will play a crucial role in detecting the most suitable candidates for immunomodulation.
Collapse
Affiliation(s)
- Evdoxia Kyriazopoulou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | | |
Collapse
|
29
|
Paranga TG, Pavel-Tanasa M, Constantinescu D, Plesca CE, Petrovici C, Miftode IL, Moscalu M, Cianga P, Miftode EG. Comparison of C-reactive protein with distinct hyperinflammatory biomarkers in association with COVID-19 severity, mortality and SARS-CoV-2 variants. Front Immunol 2023; 14:1213246. [PMID: 37388734 PMCID: PMC10302717 DOI: 10.3389/fimmu.2023.1213246] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/30/2023] [Indexed: 07/01/2023] Open
Abstract
C-reactive protein (CRP) has been one of the most investigated inflammatory-biomarkers during the ongoing COVID-19 pandemics caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The severe outcome among patients with SARS-CoV-2 infection is closely related to the cytokine storm and the hyperinflammation responsible for the acute respiratory distress syndrome and multiple organ failure. It still remains a challenge to determine which of the hyperinflammatory biomarkers and cytokines are the best predictors for disease severity and mortality in COVID-19 patients. Therefore, we evaluated and compared the outcome prediction efficiencies between CRP, the recently reported inflammatory modulators (suPAR, sTREM-1, HGF), and the classical biomarkers (MCP-1, IL-1β, IL-6, NLR, PLR, ESR, ferritin, fibrinogen, and LDH) in patients confirmed with SARS-CoV-2 infection at hospital admission. Notably, patients with severe disease had higher serum levels of CRP, suPAR, sTREM-1, HGF and classical biomarkers compared to the mild and moderate cases. Our data also identified CRP, among all investigated analytes, to best discriminate between severe and non-severe forms of disease, while LDH, sTREM-1 and HGF proved to be excellent mortality predictors in COVID-19 patients. Importantly, suPAR emerged as a key molecule in characterizing the Delta variant infections.
Collapse
Affiliation(s)
- Tudorita Gabriela Paranga
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- St. Parascheva Clinical Hospital for Infectious Diseases, Iasi, Romania
| | - Mariana Pavel-Tanasa
- Department of Immunology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Laboratory of Immunology, St. Spiridon County Clinical Emergency Hospital, Iasi, Romania
| | - Daniela Constantinescu
- Department of Immunology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Laboratory of Immunology, St. Spiridon County Clinical Emergency Hospital, Iasi, Romania
| | - Claudia Elena Plesca
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- St. Parascheva Clinical Hospital for Infectious Diseases, Iasi, Romania
| | - Cristina Petrovici
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- St. Parascheva Clinical Hospital for Infectious Diseases, Iasi, Romania
| | - Ionela-Larisa Miftode
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- St. Parascheva Clinical Hospital for Infectious Diseases, Iasi, Romania
| | - Mihaela Moscalu
- Department of Preventive Medicine and Interdisciplinarity, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Petru Cianga
- Department of Immunology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Laboratory of Immunology, St. Spiridon County Clinical Emergency Hospital, Iasi, Romania
| | - Egidia Gabriela Miftode
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- St. Parascheva Clinical Hospital for Infectious Diseases, Iasi, Romania
| |
Collapse
|
30
|
Fu Y, Xue H, Wang T, Ding Y, Cui Y, Nie H. Fibrinolytic system and COVID-19: From an innovative view of epithelial ion transport. Biomed Pharmacother 2023; 163:114863. [PMID: 37172333 PMCID: PMC10169260 DOI: 10.1016/j.biopha.2023.114863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/14/2023] Open
Abstract
Lifeways of worldwide people have changed dramatically amid the coronavirus disease 2019 (COVID-19) pandemic, and public health is at stake currently. In the early stage of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, fibrinolytic system is mostly inhibited, which is responsible for the development of hypofibrinolysis, promoting disseminated intravascular coagulation, hyaline membrane formation, and pulmonary edema. Whereas the common feature and risk factor at advanced stage is a large amount of fibrin degradation products, including D-dimer, the characteristic of hyperfibrinolysis. Plasmin can cleave both SARS-CoV-2 spike protein and γ subunit of epithelial sodium channel (ENaC), a critical element to edematous fluid clearance. In this review, we aim to sort out the role of fibrinolytic system in the pathogenesis of COVID-19, as well as provide the possible guidance in current treating methods. In addition, the abnormal regulation of ENaC in the occurrence of SARS-CoV-2 mediated hypofibrinolysis and hyperfibrinolysis are summarized, with the view of proposing an innovative view of epithelial ion transport in preventing the dysfunction of fibrinolytic system during the progress of COVID-19.
Collapse
Affiliation(s)
- Yunmei Fu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Hao Xue
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Tingyu Wang
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yong Cui
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang 110001, China.
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China.
| |
Collapse
|
31
|
Vassiliou AG, Vrettou CS, Keskinidou C, Dimopoulou I, Kotanidou A, Orfanos SE. Endotheliopathy in Acute COVID-19 and Long COVID. Int J Mol Sci 2023; 24:8237. [PMID: 37175942 PMCID: PMC10179170 DOI: 10.3390/ijms24098237] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/28/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
The pulmonary endothelium is a highly regulated organ that performs a wide range of functions under physiological and pathological conditions. Since endothelial dysfunction has been demonstrated to play a direct role in sepsis and acute respiratory distress syndrome, its role in COVID-19 has also been extensively investigated. Indeed, apart from the COVID-19-associated coagulopathy biomarkers, new biomarkers were recognised early during the pandemic, including markers of endothelial cell activation or injury. We systematically searched the literature up to 10 March 2023 for studies examining the association between acute and long COVID-19 severity and outcomes and endothelial biomarkers.
Collapse
Affiliation(s)
- Alice G. Vassiliou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (C.S.V.); (C.K.); (I.D.); (A.K.)
| | | | | | | | | | - Stylianos E. Orfanos
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (C.S.V.); (C.K.); (I.D.); (A.K.)
| |
Collapse
|
32
|
Abdellatif HAA, Sultan BO, Nassar HM, Gomaa MEE, Sakr MG, Riad E, Al-Harbi AI, Abdulhakim JA, Fawzy MS, Abd El-Fadeal NM. Circulating Soluble Urokinase Plasminogen Activator Receptor as a Predictive Indicator for COVID-19-Associated Acute Kidney Injury and Mortality: Clinical and Bioinformatics Analysis. Int J Mol Sci 2023; 24:7177. [PMID: 37108340 PMCID: PMC10138406 DOI: 10.3390/ijms24087177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Urokinase receptors regulate the interplay between inflammation, immunity, and blood clotting. The soluble urokinase plasminogen activator system is an immunologic regulator affecting endothelial function and its related receptor; the soluble urokinase plasminogen activator receptor (suPAR) has been reported to impact kidney injury. This work aims to measure serum levels of suPAR in COVID-19 patients and correlate the measurements with variable clinicolaboratory parameters and patient outcomes. In this prospective cohort study, 150 COVID-19 patients and 50 controls were included. The circulating suPAR levels were quantified by Enzyme-linked immunosorbent assay (ELISA). Routine COVID-19 laboratory assessments, including CBC, CRP, LDH, serum creatinine, and estimated glomerular filtration rates, were performed. The need for oxygen therapy, CO-RAD score, and survival rates was assessed. Bioinformatic analysis and molecular docking were run to explore the urokinase receptor structure/function and to characterize molecules as potential anti-suPAR therapeutic targets, respectively. We found higher circulating suPAR levels in COVID-19 patients vs. controls (p < 0.001). Circulating suPAR levels positively correlated with COVID-19 severity, the need for O2 therapy, the total leukocytes count, and the neutrophils to lymphocyte ratio, while they were negatively correlated with the O2 saturation level, albumin, blood calcium, lymphocytic count, and GFR. In addition, the suPAR levels were associated with poor prognostic outcomes such as a high incidence of acute kidney injury (AKI) and mortality rate. Kaplan-Meier curves showed a lower survival rate with higher suPAR levels. The logistic regression analysis confirmed the significant association of suPAR levels with the occurrence of AKI related to COVID-19 and with increased mortality probability within three months of COVID-19 follow-up. Some compounds that can act similarly to uPAR were discovered and tested by molecular docking to identify the possible ligand-protein interactions. In conclusion, higher circulating suPAR levels were associated with COVID-19 severity and could be considered a putative predictor of AKI development and mortality.
Collapse
Affiliation(s)
- Hidi A. A. Abdellatif
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Oncology Diagnostic Unit, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Basma Osman Sultan
- Internal Medicine Department-Nephrology Unit, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Hassnaa M. Nassar
- Clinical Pathology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | | | - Mohamed Gamal Sakr
- Internal Medicine Department-Tropical Medicine, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Eman Riad
- Pulmonology Unit, Internal Medicine Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Alhanouf I. Al-Harbi
- Department of Medical Laboratory, College of Applied Medical Sciences, Taibah University, Yanbu 46411, Saudi Arabia
| | - Jawaher A. Abdulhakim
- Department of Medical Laboratory, College of Applied Medical Sciences, Taibah University, Yanbu 46411, Saudi Arabia
| | - Manal S. Fawzy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 1321, Saudi Arabia
| | - Noha M. Abd El-Fadeal
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Oncology Diagnostic Unit, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Center of Excellence in Molecular and Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
33
|
Nusshag C, Wei C, Hahm E, Hayek SS, Li J, Samelko B, Rupp C, Szudarek R, Speer C, Kälble F, Schaier M, Uhle F, Schmitt FC, Fiedler MO, Krautkrämer E, Cao Y, Rodriguez R, Merle U, Eugen-Olsen J, Zeier M, Weigand MA, Morath C, Brenner T, Reiser J. suPAR links a dysregulated immune response to tissue inflammation and sepsis-induced acute kidney injury. JCI Insight 2023; 8:165740. [PMID: 37036003 PMCID: PMC10132159 DOI: 10.1172/jci.insight.165740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/21/2023] [Indexed: 04/11/2023] Open
Abstract
Acute kidney injury (AKI) secondary to sepsis results in poor outcomes and conventional kidney function indicators lack diagnostic value. Soluble urokinase plasminogen activator receptor (suPAR) is an innate immune-derived molecule implicated in inflammatory organ damage. We characterized the diagnostic ability of longitudinal serum suPAR levels to discriminate severity and course of sepsis-induced AKI (SI-AKI) in 200 critically ill patients meeting Sepsis-3 criteria. The pathophysiologic relevance of varying suPAR levels in SI-AKI was explored in a polymicrobial sepsis model in WT, (s)uPAR-knockout, and transgenic suPAR-overexpressing mice. At all time points studied, suPAR provided a robust classification of SI-AKI disease severity, with improved prediction of renal replacement therapy (RRT) and mortality compared with established kidney biomarkers. Patients with suPAR levels of greater than 12.7 ng/mL were at highest risk for RRT or death, with an adjusted odds ratio of 7.48 (95% CI, 3.00-18.63). suPAR deficiency protected mice against SI-AKI. suPAR-overexpressing mice exhibited greater kidney damage and poorer survival through inflamed kidneys, accompanied by local upregulation of potent chemoattractants and pronounced kidney T cell infiltration. Hence, suPAR allows for an innate immune-derived and kidney function-independent staging of SI-AKI and offers improved longitudinal risk stratification. suPAR promotes T cell-based kidney inflammation, while suPAR deficiency improves SI-AKI.
Collapse
Affiliation(s)
- Christian Nusshag
- Department of Internal Medicine, RUSH University Medical Center, Chicago, Illinois, USA
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Changli Wei
- Department of Internal Medicine, RUSH University Medical Center, Chicago, Illinois, USA
| | - Eunsil Hahm
- Department of Internal Medicine, RUSH University Medical Center, Chicago, Illinois, USA
| | - Salim S Hayek
- Department of Medicine, Division of Cardiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jing Li
- Department of Internal Medicine, RUSH University Medical Center, Chicago, Illinois, USA
| | - Beata Samelko
- Department of Internal Medicine, RUSH University Medical Center, Chicago, Illinois, USA
| | | | | | - Claudius Speer
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian Kälble
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Matthias Schaier
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | | | | | | | - Ellen Krautkrämer
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Yanxia Cao
- Department of Internal Medicine, RUSH University Medical Center, Chicago, Illinois, USA
| | - Ricardo Rodriguez
- Department of Internal Medicine, RUSH University Medical Center, Chicago, Illinois, USA
| | - Uta Merle
- Department of Gastroenterology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Martin Zeier
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Christian Morath
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thorsten Brenner
- Department of Anesthesiology, and
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jochen Reiser
- Department of Internal Medicine, RUSH University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
34
|
Zanelli S, Bakakos A, Sotiropoulou Z, Papaioannou AI, Koukaki E, Potamianou E, Kyriakoudi A, Kaniaris E, Bakakos P, Giamarellos-Bourboulis EJ, Koutsoukou A, Rovina N. Modified SCOPE (mSCOPE) Score as a Tool to Predict Mortality in COVID-19 Critically Ill Patients. J Pers Med 2023; 13:jpm13040628. [PMID: 37109014 PMCID: PMC10146219 DOI: 10.3390/jpm13040628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Introduction: Efficient clinical scores predicting the outcome of severe COVID-19 pneumonia may play a pivotal role in patients’ management. The aim of this study was to assess the modified Severe COvid Prediction Estimate score (mSCOPE) index as a predictor of mortality in patients admitted to the ICU due to severe COVID-19 pneumonia. Materials and methods: In this retrospective observational study, 268 critically ill COVID-19 patients were included. Demographic and laboratory characteristics, comorbidities, disease severity, and outcome were retrieved from the electronical medical files. The mSCOPE was also calculated. Results: An amount of 70 (26.1%) of patients died in the ICU. These patients had higher mSCOPE score compared to patients who survived (p < 0.001). mSCOPE correlated to disease severity (p < 0.001) and to the number and severity of comorbidities (p < 0.001). Furthermore, mSCOPE significantly correlated with days on mechanical ventilation (p < 0.001) and days of ICU stay (p = 0.003). mSCOPE was found to be an independent predictor of mortality (HR:1.219, 95% CI: 1.010–1.471, p = 0.039), with a value ≥ 6 predicting poor outcome with a sensitivity (95%CI) 88.6%, specificity 29.7%, a positive predictive value of 31.5%, and a negative predictive value of 87.7%. Conclusion: mSCOPE score could be proved useful in patients’ risk stratification, guiding clinical interventions in patients with severe COVID-19.
Collapse
Affiliation(s)
- Stavroula Zanelli
- 1st Respiratory Department, Medical School, National and Kapodistrian University of Athens, “Sotiria” Chest Hospital, 11527 Athens, Greece
| | - Agamemnon Bakakos
- 1st Respiratory Department, Medical School, National and Kapodistrian University of Athens, “Sotiria” Chest Hospital, 11527 Athens, Greece
| | - Zoi Sotiropoulou
- 1st Respiratory Department, Medical School, National and Kapodistrian University of Athens, “Sotiria” Chest Hospital, 11527 Athens, Greece
| | - Andriana I. Papaioannou
- 1st Respiratory Department, Medical School, National and Kapodistrian University of Athens, “Sotiria” Chest Hospital, 11527 Athens, Greece
| | - Evangelia Koukaki
- 1st Respiratory Department, Medical School, National and Kapodistrian University of Athens, “Sotiria” Chest Hospital, 11527 Athens, Greece
| | - Efstathia Potamianou
- 1st Respiratory Department, Medical School, National and Kapodistrian University of Athens, “Sotiria” Chest Hospital, 11527 Athens, Greece
| | - Anna Kyriakoudi
- 1st Respiratory Department, Medical School, National and Kapodistrian University of Athens, “Sotiria” Chest Hospital, 11527 Athens, Greece
| | - Evangelos Kaniaris
- 1st Respiratory Department, Medical School, National and Kapodistrian University of Athens, “Sotiria” Chest Hospital, 11527 Athens, Greece
| | - Petros Bakakos
- 1st Respiratory Department, Medical School, National and Kapodistrian University of Athens, “Sotiria” Chest Hospital, 11527 Athens, Greece
| | - Evangelos J. Giamarellos-Bourboulis
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, “Attikon” Hospital, 12462 Athens, Greece
| | - Antonia Koutsoukou
- 1st Respiratory Department, Medical School, National and Kapodistrian University of Athens, “Sotiria” Chest Hospital, 11527 Athens, Greece
| | - Nikoletta Rovina
- 1st Respiratory Department, Medical School, National and Kapodistrian University of Athens, “Sotiria” Chest Hospital, 11527 Athens, Greece
| |
Collapse
|
35
|
Bacharaki D, Petrakis I, Stylianou K. Redefying the therapeutic strategies against cardiorenal morbidity and mortality: Patient phenotypes. World J Cardiol 2023; 15:76-83. [PMID: 37033683 PMCID: PMC10074996 DOI: 10.4330/wjc.v15.i3.76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/31/2022] [Accepted: 02/22/2023] [Indexed: 03/21/2023] Open
Abstract
Chronic kidney disease (CKD) patients face an unacceptably high morbidity and mortality, mainly from cardiovascular diseases. Diabetes mellitus, arterial hypertension and dyslipidemia are highly prevalent in CKD patients. Established therapeutic protocols for the treatment of diabetes mellitus, arterial hypertension, and dyslipidemia are not as effective in CKD patients as in the general population. The role of non-traditional risk factors (RF) has gained interest in the last decades. These entail the deranged clinical spectrum of secondary hyperparathyroidism involving vascular and valvular calcification, under the term "CKD-mineral and bone disorder" (CKD-MBD), uremia per se, inflammation and oxidative stress. Each one of these non-traditional RF have been addressed in various study designs, but the results do not exhibit any applied clinical benefit for CKD-patients. The "crusade" against cardiorenal morbidity and mortality in CKD-patients is in some instances, derailed. We propose a therapeutic paradigm advancing from isolated treatment targets, as practiced today, to precision medicine involving patient phenotypes with distinct underlying pathophysiology. In this regard we propose two steps, based on current stratification management of corona virus disease-19 and sepsis. First, select patients who are expected to have a high mortality, i.e., a prognostic enrichment. Second, select patients who are likely to respond to a specific therapy, i.e., a predictive enrichment.
Collapse
Affiliation(s)
- Dimitra Bacharaki
- Nephrology Unit, 2 Department of Internal Medicine, Attikon University Hospital, Chaidari 12462, Greece.
| | - Ioannis Petrakis
- Department of Nephrology, Heraklion University Hospital, University of Crete, Heraklion 71500, Greece
| | - Kostas Stylianou
- Department of Nephrology, Heraklion University Hospital, University of Crete, Heraklion 71500, Greece
| |
Collapse
|
36
|
Aomar-Millán IF, de Victoria-Carazo JM, Fernández Reyes D, Torres-Parejo Ú, Pérez Fernández L, Martínez-Diz S, Ceballos Torres A, López Gómez J, Bizzarri F, Raya Álvarez E, Salvatierra J. Characteristics and clinical outcome in 312 patients with moderate to severe pneumonia due to SARS-COV-2 and hyperinflammation treated with anakinra and corticosteroids: A retrospective cohort study. PLoS One 2023; 18:e0283529. [PMID: 36961847 PMCID: PMC10038301 DOI: 10.1371/journal.pone.0283529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/12/2023] [Indexed: 03/25/2023] Open
Abstract
OBJECTIVE To assess the clinical outcome (death and/or Intensive Care Unit (ICU) admission) based on the time from hospital admission to the administration of anakinra and the possible usefulness of a "simplified" SCOPE score to stratify the risk of worse prognosis in our cohort of patients with moderate/severe SARS-CoV-2 pneumonia, both vaccinated and unvaccinated, that received anakinra and corticosteroids. In addition, the clinical, analytical, and imaging characteristics of patients at admission are described. METHODS Retrospective cohort study of 312 patients admitted to Hospital Clínico San Cecilio in Granada for moderate/severe pneumonia caused by SARS-CoV-2 that received anakinra and corticosteroids between March 2020 and January 2022. Clinical and analytical data were collected as well as the patient outcome at 30 and 60 days after admission. Three treatment groups were established according to the time from hospital admission to administration of anakinra: early (1st-2nd day), intermediate (3rd-5th day), and late (after the 5th day). RESULTS The median age was 67.4 years (IQR 22-97 years) and 204 (65.4%) were male. The most common comorbidity was hypertension (58%). The median time from the start of symptoms to anakinra administration was 6 days (IQR 5-10) and the SaFi (SaO2/FiO2) was 228 (IQR 71-471). The cure rate was higher in the early-onset anakinra group versus the late-onset group (73% vs 56.6%). The latter had a higher percentage of deaths (27.4%) and a greater number of patients remained hospitalized for a month (16%). On admission, the patients had elevated C-reactive protein (CRP), ferritin, and D-dimer values and decreased total lymphocytes. Analytical improvement was observed at both 72 hours and one month after treatment. 42 (13.5%) required ICU admission, and 23 (7.3%) orotracheal intubation. At 60 days, 221 (70.8%) were discharged, 87 (27.8%) had died and 4 (1.4%) remained hospitalized. The mean dose of anakinra was 1000 mg (100-2600 mg) with differences found between the dose administered and the clinical outcome. There were no differences in the primary outcome based on vaccination. A simplified SCOPE score at the start of anakinra administration was lower in patients with better clinical evolution. CONCLUSIONS Early treatment with anakinra and corticosteroids was associated with a better outcome regardless of vaccination status. A simplified SCOPE was found to be a good prognostic tool.
Collapse
Affiliation(s)
- Ismael Francisco Aomar-Millán
- Department of Internal Medicine, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Biosanitary Research Institute of Granada, ibsGRANADA, Granada, Spain
| | | | - Daniel Fernández Reyes
- Department of Internal Medicine, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Úrsula Torres-Parejo
- Department of Statistics, Faculty of Health Sciences, University of Granada, Granada, Spain
| | | | - Silvia Martínez-Diz
- Department of Preventive Medicine, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Angel Ceballos Torres
- Department of Internal Medicine, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Jairo López Gómez
- Department of Internal Medicine, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Francesco Bizzarri
- Department of Rheumatology, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Enrique Raya Álvarez
- Biosanitary Research Institute of Granada, ibsGRANADA, Granada, Spain
- Department of Rheumatology, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Juan Salvatierra
- Biosanitary Research Institute of Granada, ibsGRANADA, Granada, Spain
- Department of Rheumatology, Hospital Universitario Clínico San Cecilio, Granada, Spain
| |
Collapse
|
37
|
Cavaillon JM, Artigas A, Barratt-Due A, Giamarellos-Bourboulis EJ, Gómez H, Hayem G, Vlaar APJ, Wiersinga WJ. SEVERE CORONAVIRUS DISEASE 2019: FROM PATHOGENESIS TO THERAPY. Shock 2023; 59:10-15. [PMID: 36469709 DOI: 10.1097/shk.0000000000001956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
ABSTRACT The COVID-19 pandemic has been a challenge to propose efficient therapies. Because severe SARS-CoV2 infection is a viral sepsis eventually followed by an immunological autoinflammatory phenomenon, many approaches have been inspired by the previous attempts made in bacterial sepsis, while specific antiviral strategies (use of interferon or specific drugs) have been additionally investigated. We summarize our current thinking on the use of SARS-CoV-2 antivirals, corticosteroids, anti-IL-1, anti-IL-6, anti-C5a, as well as stem cell therapy in severe COVID-19. Patient stratification and appropriate time window will be important to be defined to guide successful treatment.
Collapse
Affiliation(s)
| | - Antonio Artigas
- Intensive Care Department, Corporacion Sanitaria Universitaria Parc Tauli CIBER Enfermedades Respiratorias, Autonomous University of Barcelona, Sabadell, Spain
| | | | | | - Hernando Gómez
- Program for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh Medical Center, Pennsylvania, PA
| | - Gilles Hayem
- Rheumatology Department, Paris Saint-Joseph Hospital, Paris, France
| | - Alexander P J Vlaar
- Department of Intensive Care, Amsterdam University Medical Center, University of Amsterdam, the Netherlands
| | - W Joost Wiersinga
- Division of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
38
|
Tsangaris I, Antonakos N, Fantoni M, Kaplanski G, Kyriazopoulou E, Veas F, Clemens M. BIOMARKERS: CAN THEY REALLY GUIDE OUR DAILY PRACTICE? Shock 2023; 59:16-20. [PMID: 36867757 DOI: 10.1097/shk.0000000000001957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
ABSTRACT Optimal management of septic patients requires accurate assessment of both current severity status and prognosis. Since the 1990s, substantial advances have been made in the use of circulating biomarkers for such assessments. This summary of the session on "Biomarkers: can they really use guide our daily practice?" presented at the 2021 WEB-CONFERENCE OF THE EUROPEAN SHOCK SOCIETY, 6 November 2021. These biomarkers include ultrasensitive detection of bacteremia, circulating soluble urokina-type plasminogen activator receptor (suPAR), C-reactive protein (CRP) and ferritin and procalcitonin. In addition, the potential application of novel multiwavelength optical biosensor technology allows noninvasive monitoring of multiple metabolites that can be used to assess severity and prognosis in septic patients. The application these biomarkers and improved technologies provide the potential for improved personalized management of septic patients.
Collapse
Affiliation(s)
- Iraklis Tsangaris
- 2nd Department of Critical Care Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Antonakos
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Gilles Kaplanski
- Division of Internal Medicine and Clinical Immunology, Hôpital de la Conception, C2VN-INSERM U1263, Aix-Marseille University, Marseille, France
| | - Evdoxia Kyriazopoulou
- 2nd Department of Critical Care Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Mark Clemens
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina
| |
Collapse
|
39
|
Akinosoglou K, Kotsaki A, Gounaridi IM, Christaki E, Metallidis S, Adamis G, Fragkou A, Fantoni M, Rapti A, Kalomenidis I, Chrysos G, Boni G, Kainis I, Alexiou Z, Castelli F, Serino FS, Bakakos P, Nicastri E, Tzavara V, Safarika A, Ioannou S, Dagna L, Dimakou K, Tzatzagou G, Chini M, Bassetti M, Kotsis V, Angheben A, Tsoukalas G, Selmi C, Spiropoulou OM, Samarkos M, Doumas M, Damoraki G, Masgala A, Papanikolaou I, Argyraki A, Negri M, Leventogiannis K, Sympardi S, Gatselis NK, Petrakis V, Netea MG, Panagopoulos P, Sakka V, Milionis H, Dalekos GN, Giamarellos-Bourboulis EJ. Efficacy and safety of early soluble urokinase plasminogen receptor plasma-guided anakinra treatment of COVID-19 pneumonia: A subgroup analysis of the SAVE-MORE randomised trial. EClinicalMedicine 2023; 56:101785. [PMID: 36590789 PMCID: PMC9791950 DOI: 10.1016/j.eclinm.2022.101785] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/27/2022] Open
Abstract
Background The SAVE-MORE trial demonstrated that anakinra treatment in COVID-19 pneumonia with plasma soluble urokinase plasminogen activator (suPAR) levels of 6 ng/mL or more was associated with 0.36 odds for a worse outcome compared to placebo when expressed by the WHO-Clinical Progression Scale (CPS) at day 28. Herein, we report the results of subgroup analyses and long-term outcomes. Methods This prospective, double-blind, randomised clinical trial, recruited patients with a confirmed SARS-CoV-2 infection, in need of hospitalisation, lower respiratory tract infection and plasma suPAR ≥6 ng/mL from 37 academic and community hospitals in Greece and Italy. Patients were 1:2 randomised to subcutaneous treatment with placebo or anakinra (100 mg) once daily for 10 days. Pre-defined subgroups of Charlson's comorbidity index (CCI), sex, age, level of suPAR, and time from symptom onset were analysed for the primary endpoint (overall comparison of distribution of frequencies of the scores from the WHO-CPS between treatments on day 28), by multivariable ordinal regression analysis in the intention to treat (ITT) population. This trial is registered with the EU Clinical Trials Register (2020-005828-11) and ClinicalTrials.gov (NCT04680949). Findings Patients were enrolled between 23 December 2020 and 31 March 2021; 189 patients in the placebo arm and 405 patients in the anakinra arm were the ITT population. Multivariable analysis showed that anakinra treatment was accompanied by significantly lower odds for worse outcome compared to placebo at day 28 for all studied subgroups (CCI ≥ 2, OR: 0.34, 95% confidence intervals [CI] 0.22-0.50; CCI < 2, OR: 0.38, 95% CI 0.21-0.68; suPAR > 9 ng/mL, OR: 0.35, 95% CI 0.19-0.66; suPAR 6-9 ng/mL, OR: 0.35, 95% CI 0.24-0.52; patients ≥65 years, OR: 0.41, 95% CI 0.25-0.66; and patients <65 years, OR: 0.29, 95% CI 0.19-0.45). The benefit was uniform, irrespective of the time from start of symptoms until the start of the study drug. At days 60 and 90, anakinra treatment had odds of 0.40 (95% CI 0.28-0.57) and 0.46 (95% CI 0.32-0.67) respectively, for a worse outcome compared to placebo. The costs of general ward stay, ICU stay, and drugs were lower with anakinra treatment. Interpretation Anakinra represents an important therapeutic tool in the management of COVID-19 that may be administered in all subgroups of patients; benefits are maintained until day 90. Funding Hellenic Institute for the Study of Sepsis; Swedish Orphan Biovitrum AB.
Collapse
Affiliation(s)
| | - Antigone Kotsaki
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Hellenic Institute for the Study of Sepsis, Athens, Greece
| | - Ioanna-Maria Gounaridi
- Third Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Eirini Christaki
- First Department of Internal Medicine, Medical School, University of Ioannina, Ioannina, Greece
| | - Simeon Metallidis
- First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Adamis
- First Department of Internal Medicine, G. Gennimatas General Hospital of Athens, Athens, Greece
| | | | - Massimo Fantoni
- Dipartimento Scienze di Laboratorio e Infettivologiche - Fondazione Policlinico Gemelli IRCCS, Roma, Italy
| | - Aggeliki Rapti
- Second Department of Pulmonary Medicine, Sotiria General Hospital of Chest Diseases, Athens, Greece
| | - Ioannis Kalomenidis
- First Department of Critical Care and Pulmonary Medicine, Medical School, Evangelismos General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Chrysos
- Second Department of Internal Medicine, Tzaneio General Hospital of Piraeus, Athens, Greece
| | - Gloria Boni
- Hospital Pharmacy, IRCCS Hospital Sacro Cuore, Negrar di Valpolicella, Verona, Italy
- IRCSS Sacro Cuore Hospital, Negrar di Valpolicella, Verona, Italy
| | - Ilias Kainis
- Tenth Department of Pulmonary Medicine, Sotiria General Hospital of Chest Diseases of Athens, Athens, Greece
| | - Zoi Alexiou
- Second Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Francesco Castelli
- Spedali Civili, Brescia ASST Spedali Civili Hospital, University of Brescia, Italy
| | | | - Petros Bakakos
- First Department of Chest Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Emanuele Nicastri
- Department of Internal Medicine, Spallanzani Institute of Rome, Rome, Italy
| | - Vassiliki Tzavara
- First Department of Internal Medicine, Korgialeneion-Benakeion General Hospital, Athens, Greece
| | - Asimina Safarika
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Hellenic Institute for the Study of Sepsis, Athens, Greece
| | - Sofia Ioannou
- Department of Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS Ospedale San Raffaele & Vita-Salute San Raffaele University, Milan, Italy
| | - Katerina Dimakou
- Fifth Department of Pulmonary Medicine, Sotiria General Hospital of Chest Diseases, Athens, Greece
| | - Glykeria Tzatzagou
- First Department of Internal Medicine, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Maria Chini
- Third Department of Internal Medicine and Infectious Diseases Unit, Korgialeneion-Benakeion General Hospital, Athens, Greece
| | - Matteo Bassetti
- Infectious Diseases Clinic, Ospedale Policlinico San Martino IRCCS and Department of Health Sciences, University of Genova, Genoa, Italy
| | - Vasileios Kotsis
- Third Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Andrea Angheben
- Department of Infectious – Tropical Diseases and Microbiology, IRCSS Sacro Cuore Hospital, Negrar di Valpolicella, Verona, Italy
| | - George Tsoukalas
- Fourth Department of Pulmonary Medicine, Sotiria General Hospital of Chest Diseases, Athens, Greece
| | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele 20072, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Olga-Maria Spiropoulou
- First Department of Internal Medicine, Asklipieio General Hospital of Voula, Athens, Greece
| | - Michael Samarkos
- First Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael Doumas
- Second Department of Propedeutic Medicine, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgia Damoraki
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Masgala
- Second Department of Internal Medicine, Konstantopouleio General Hospital, Athens, Greece
| | - Ilias Papanikolaou
- Department of Pulmonary Medicine, General Hospital of Kerkyra, Kerkyra, Greece
| | - Aikaterini Argyraki
- Department of Internal Medicine, Sotiria General Hospital of Chest Diseases, Athens, Greece
| | - Marcantonio Negri
- Dipartimento Scienze Mediche e Chirurgiche - Fondazione Policlinico Gemelli IRCCS, Roma, Italy
| | - Konstantinos Leventogiannis
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Hellenic Institute for the Study of Sepsis, Athens, Greece
| | - Styliani Sympardi
- First Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Nikolaos K. Gatselis
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, Full Member of the European Reference Network on Hepatological Disases (ERN RARE-LIVER), General University Hospital of Larissa, Larissa 41110, Greece
| | - Vasileios Petrakis
- Second Department of Internal Medicine, Medical School, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Mihai G. Netea
- Department of Internal Medicine and Center for Infectious Diseases, Radboud University, Nijmegen 6500, Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Periklis Panagopoulos
- Second Department of Internal Medicine, Medical School, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Vissaria Sakka
- Third Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Haralampos Milionis
- First Department of Internal Medicine, Medical School, University of Ioannina, Ioannina, Greece
| | - George N. Dalekos
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, Full Member of the European Reference Network on Hepatological Disases (ERN RARE-LIVER), General University Hospital of Larissa, Larissa 41110, Greece
| | - Evangelos J. Giamarellos-Bourboulis
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Hellenic Institute for the Study of Sepsis, Athens, Greece
| |
Collapse
|
40
|
Samaras C, Kyriazopoulou E, Poulakou G, Reiner E, Kosmidou M, Karanika I, Petrakis V, Adamis G, Gatselis NK, Fragkou A, Rapti A, Taddei E, Kalomenidis I, Chrysos G, Bertoli G, Kainis I, Alexiou Z, Castelli F, Saverio Serino F, Bakakos P, Nicastri E, Tzavara V, Kostis E, Dagna L, Koukidou S, Tzatzagou G, Chini M, Bassetti M, Trakatelli C, Tsoukalas G, Selmi C, Samarkos M, Pyrpasopoulou A, Masgala A, Antonakis E, Argyraki A, Akinosoglou K, Sympardi S, Panagopoulos P, Milionis H, Metallidis S, Syrigos KN, Angel A, Dalekos GN, Netea MG, Giamarellos-Bourboulis EJ. Interferon gamma-induced protein 10 (IP-10) for the early prognosis of the risk for severe respiratory failure and death in COVID-19 pneumonia. Cytokine 2023; 162:156111. [PMID: 36529030 PMCID: PMC9747699 DOI: 10.1016/j.cyto.2022.156111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/18/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Elevated concentrations of soluble urokinase plasminogen activator receptor (suPAR) predict progression to severe respiratory failure (SRF) or death among patients with COVID-19 pneumonia and guide early anakinra treatment. As suPAR testing may not be routinely available in every health-care setting, alternative biomarkers are needed. We investigated the performance of C-reactive protein (CRP), interferon gamma-induced protein-10 (IP-10) and TNF-related apoptosis-inducing ligand (TRAIL) for predicting SRF or death in COVID-19. METHODS Two cohorts were studied; one discovery cohort with 534 patients from the SAVE-MORE clinical trial; and one validation cohort with 364 patients from the SAVE trial including also 145 comparators. CRP, IP-10 and TRAIL were measured by the MeMed Key® platform in order to select the biomarker with the best prognostic performance for the early prediction of progression into SRF or death. RESULTS IP-10 had the best prognostic performance: baseline concentrations 2000 pg/ml or higher predicted equally well to suPAR (sensitivity 85.0 %; negative predictive value 96.6 %). Odds ratio for poor outcome among anakinra-treated participants of the SAVE-MORE trial was 0.35 compared to placebo when IP-10 was 2,000 pg/ml or more. IP-10 could divide different strata of severity for SRF/death by day 14 in the validation cohort. Anakinra treatment decreased this risk irrespective the IP-10 concentrations. CONCLUSIONS IP-10 concentrations of 2,000 pg/ml or higher are a valid alternative to suPAR for the early prediction of progression into SRF or death the first 14 days from hospital admission for COVID-19 and they may guide anakinra treatment. TRIAL REGISTRATION CLINICALTRIALS gov, NCT04680949 and NCT04357366.
Collapse
Affiliation(s)
- Charilaos Samaras
- 1(st) Department of Internal Medicine, Asklepieio General Hospital of Voula, Greece
| | - Evdoxia Kyriazopoulou
- 4(th) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece; Hellenic Institute for the Study of Sepsis, Athens, Greece
| | - Garyfallia Poulakou
- 3(rd) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - Maria Kosmidou
- 1st Department of Internal Medicine, University of Ioannina, Medical School, Ioannina, Greece
| | - Ioanna Karanika
- 1st Department of Internal Medicine, Aristotle University of Thessaloniki, Medical School, Thessaloniki, Greece
| | - Vasileios Petrakis
- 2(nd) Department of Internal Medicine, Democritus University of Thrace, Medical School, 681 00, Alexandroupolis, Greece
| | - George Adamis
- 1(st) Department of Internal Medicine, G. Gennimatas General Hospital of Athens, Athens, Greece
| | - Nikolaos K Gatselis
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, Full Member of the European Reference Network on Hepatological Disases (ERN RARE-LIVER), General University Hospital of Larissa, 41110 Larissa, Greece
| | | | - Aggeliki Rapti
- 2(nd) Department of Pulmonary Medicine, Sotiria General Hospital of Chest Diseases, Athens, Greece
| | - Eleonora Taddei
- Dipartimento Scienze di Laboratorio e Infettivologiche - Fondazione Policlinico Universitario Agostino Gemelli IRCCS - Roma, Italy
| | - Ioannis Kalomenidis
- 1(st) Department of Critical Care and Pulmonary Medicine, Medical School, National and Kapodistrian University of Athens, Evangelismos General Hospital, Athens, Greece
| | - George Chrysos
- 2(nd) Department of Internal Medicine, Tzaneio General Hospital of Piraeus, Athens, Greece
| | - Giulia Bertoli
- Department of Infectious - Tropical Diseases and Microbiology, IRCSS Sacro Cuore Hospital, Negrar, Verona, Italy
| | - Ilias Kainis
- 10(th) Department of Pulmonary Medicine, Sotiria General Hospital of Chest Diseases of Athens, Greece
| | - Zoi Alexiou
- 2(nd) Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Francesco Castelli
- Spedali Civili, Brescia ASST Spedali Civili Hospital, University of Brescia, Italy
| | | | - Petros Bakakos
- 1(st) Department of Chest Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Emanuele Nicastri
- Department of Internal Medicine, Spallanzani Institute of Rome, Italy
| | - Vassiliki Tzavara
- 1(st) Department of Internal Medicine, Korgialeneion-Benakeion General Hospital, Athens, Greece
| | - Evangelos Kostis
- Department of Therapeutics, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS Ospedale San Raffaele & Vita-Salute San Raffaele University, Milan, Italy
| | - Sofia Koukidou
- 5(th) Department of Pulmonary Medicine, Sotiria General Hospital of Chest Diseases, Athens, Greece
| | - Glykeria Tzatzagou
- 1(st) Department of Internal Medicine, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Maria Chini
- 3rd Dpt of Internal Medicine and Infectious Diseases Unit, Korgialeneion-Benakeion General Hospital, Athens, Greece
| | - Matteo Bassetti
- Infectious Diseases Clinic, Ospedale Policlinico San Martino IRCCS and Department of Health Sciences, University of Genova, Genova, Italy
| | - Christina Trakatelli
- 3(rd) Department of Internal Medicine, Aristotle University of Thessaloniki, Medical School, Thessaloniki, Greece
| | - George Tsoukalas
- 4(th) Department of Pulmonary Medicine, Sotiria General Hospital of Chest Diseases, Athens, Greece
| | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele and IRCCS Humanitas Research Hospital, via Manzoni, 56, 20089 Rozzano, Milan, Italy
| | - Michael Samarkos
- 1(st) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Athina Pyrpasopoulou
- 2(nd) Department of Propedeutic Medicine, Aristotle University of Thessaloniki, Medical School, Thessaloniki, Greece
| | - Aikaterini Masgala
- 2(nd) Department of Internal Medicine, Konstantopouleio General Hospital, Athens, Greece
| | | | - Aikaterini Argyraki
- Department of Internal Medicine, Sotiria General Hospital of Chest Diseases, Greece
| | | | - Styliani Sympardi
- 1(st) Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Periklis Panagopoulos
- 2(nd) Department of Internal Medicine, Democritus University of Thrace, Medical School, 681 00, Alexandroupolis, Greece
| | - Haralampos Milionis
- 1st Department of Internal Medicine, University of Ioannina, Medical School, Ioannina, Greece
| | - Simeon Metallidis
- 1st Department of Internal Medicine, Aristotle University of Thessaloniki, Medical School, Thessaloniki, Greece
| | - Konstantinos N Syrigos
- 3(rd) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - George N Dalekos
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, Full Member of the European Reference Network on Hepatological Disases (ERN RARE-LIVER), General University Hospital of Larissa, 41110 Larissa, Greece
| | - Mihai G Netea
- Department of Internal Medicine and Center for Infectious Diseases, Radboud University, Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Germany
| | - Evangelos J Giamarellos-Bourboulis
- 4(th) Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece; Hellenic Institute for the Study of Sepsis, Athens, Greece.
| |
Collapse
|
41
|
Yatsenko T, Skrypnyk M, Troyanovska O, Tobita M, Osada T, Takahashi S, Hattori K, Heissig B. The Role of the Plasminogen/Plasmin System in Inflammation of the Oral Cavity. Cells 2023; 12:cells12030445. [PMID: 36766787 PMCID: PMC9913802 DOI: 10.3390/cells12030445] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/03/2023] Open
Abstract
The oral cavity is a unique environment that consists of teeth surrounded by periodontal tissues, oral mucosae with minor salivary glands, and terminal parts of major salivary glands that open into the oral cavity. The cavity is constantly exposed to viral and microbial pathogens. Recent studies indicate that components of the plasminogen (Plg)/plasmin (Pm) system are expressed in tissues of the oral cavity, such as the salivary gland, and contribute to microbial infection and inflammation, such as periodontitis. The Plg/Pm system fulfills two major functions: (a) the destruction of fibrin deposits in the bloodstream or damaged tissues, a process called fibrinolysis, and (b) non-fibrinolytic actions that include the proteolytic modulation of proteins. One can observe both functions during inflammation. The virus that causes the coronavirus disease 2019 (COVID-19) exploits the fibrinolytic and non-fibrinolytic functions of the Plg/Pm system in the oral cavity. During COVID-19, well-established coagulopathy with the development of microthrombi requires constant activation of the fibrinolytic function. Furthermore, viral entry is modulated by receptors such as TMPRSS2, which is necessary in the oral cavity, leading to a derailed immune response that peaks in cytokine storm syndrome. This paper outlines the significance of the Plg/Pm system for infectious and inflammatory diseases that start in the oral cavity.
Collapse
Affiliation(s)
- Tetiana Yatsenko
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Maksym Skrypnyk
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Olga Troyanovska
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Morikuni Tobita
- Department of Oral and Maxillofacial Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Taro Osada
- Department of Gastroenterology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu-Shi 279-0021, Japan
| | - Satoshi Takahashi
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-Ku, Tokyo 108-8639, Japan
| | - Koichi Hattori
- Center for Genome and Regenerative Medicine, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
- Correspondence: (K.H.); (B.H.); Tel.: +81-3-3813-3111 (switchboard 2115) (B.H.)
| | - Beate Heissig
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
- Correspondence: (K.H.); (B.H.); Tel.: +81-3-3813-3111 (switchboard 2115) (B.H.)
| |
Collapse
|
42
|
Sarlo F, Urbani A, Baroni S. Urokinase-type plasminogen activator soluble receptor (suPAR) assay in clinical routine: evaluation one year after its introduction in the high automation corelab of the A. Gemelli hospital. Clin Chem Lab Med 2023; 61:e33-e35. [PMID: 36283064 DOI: 10.1515/cclm-2022-0686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/11/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Francesca Sarlo
- UOC di Chimica Biochimica e Biologia Molecolare Clinica, Fondazione Policlinico Universitario A. Gemelli I.R.C.C.S., Rome, Italy
| | - Andrea Urbani
- UOC di Chimica Biochimica e Biologia Molecolare Clinica, Fondazione Policlinico Universitario A. Gemelli I.R.C.C.S., Rome, Italy
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Silvia Baroni
- UOC di Chimica Biochimica e Biologia Molecolare Clinica, Fondazione Policlinico Universitario A. Gemelli I.R.C.C.S., Rome, Italy
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- UOS Corelab Biochimica Clinica Urgenze, Fondazione Policlinico Universitario A. Gemelli I.R.C.C.S., Rome, Italy
| |
Collapse
|
43
|
Shmakova AA, Popov VS, Romanov IP, Khabibullin NR, Sabitova NR, Karpukhina AA, Kozhevnikova YA, Kurilina EV, Tsokolaeva ZI, Klimovich PS, Rubina KA, Vassetzky YS, Semina EV. Urokinase System in Pathogenesis of Pulmonary Fibrosis: A Hidden Threat of COVID-19. Int J Mol Sci 2023; 24:ijms24021382. [PMID: 36674896 PMCID: PMC9867169 DOI: 10.3390/ijms24021382] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
Pulmonary fibrosis is a common and threatening post-COVID-19 complication with poorly resolved molecular mechanisms and no established treatment. The plasminogen activator system, including urokinase (uPA) and urokinase receptor (uPAR), is involved in the pathogenesis of COVID-19 and contributes to the development of lung injury and post-COVID-19 pulmonary fibrosis, although their cellular and molecular underpinnings still remain obscure. The aim of the current study was to assess the role of uPA and uPAR in the pathogenesis of pulmonary fibrosis. We analyzed uPA and uPAR expression in human lung tissues from COVID-19 patients with pulmonary fibrosis using single-cell RNA-seq and immunohistochemistry. We modeled lung fibrosis in Plau-/- and Plaur-/- mice upon bleomycin instillation and explored the effect of uPAR downregulation in A549 and BEAS-2B lung epithelial cells. We found that uPAR expression drastically decreased in the epithelial airway basal cells and monocyte/macrophage cells, whereas uPA accumulation significantly increased in tissue samples of COVID-19 patients. Lung injury and fibrosis in Plaur-/- vs. WT mice upon bleomycin instillation revealed that uPAR deficiency resulted in pro-fibrogenic uPA accumulation, IL-6 and ACE2 upregulation in lung tissues and was associated with severe fibrosis, weight loss and poor survival. uPAR downregulation in A549 and BEAS-2B was linked to an increased N-cadherin expression, indicating the onset of epithelial-mesenchymal transition and potentially contributing to pulmonary fibrosis. Here for the first time, we demonstrate that plasminogen treatment reversed lung fibrosis in Plaur-/- mice: the intravenous injection of 1 mg of plasminogen on the 21st day of bleomycin-induced fibrosis resulted in a more than a two-fold decrease in the area of lung fibrosis as compared to non-treated mice as evaluated by the 42nd day. The expression and function of the plasminogen activator system are dysregulated upon COVID-19 infection, leading to excessive pulmonary fibrosis and worsening the prognosis. The potential of plasminogen as a life-saving treatment for non-resolving post-COVID-19 pulmonary fibrosis warrants further investigation.
Collapse
Affiliation(s)
- Anna A. Shmakova
- Institute of Experimental Cardiology, National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Vladimir S. Popov
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Iliya P. Romanov
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | | | - Nailya R. Sabitova
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | | | | | - Ella V. Kurilina
- Institute of Experimental Cardiology, National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
| | - Zoya I. Tsokolaeva
- Institute of Experimental Cardiology, National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
| | - Polina S. Klimovich
- Institute of Experimental Cardiology, National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Kseniya A. Rubina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | | | - Ekaterina V. Semina
- Institute of Experimental Cardiology, National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
- Correspondence:
| |
Collapse
|
44
|
Hamza M, Alhujaily M, Alosaimi B, El Bakkouri K, AlDughaim MS, Alonazi M, Alanazi MA, Abbass B, Alshehri A, Al-Shouli ST, Alturaiki W, Awadalla M. Association between inflammatory cytokines/chemokines, clinical laboratory parameters, disease severity and in-hospital mortality in critical and mild COVID-19 patients without comorbidities or immune-mediated diseases. J Immunoassay Immunochem 2023; 44:13-30. [PMID: 35915975 DOI: 10.1080/15321819.2022.2104124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
There are limited data on inflammatory cytokines and chemokines; the humoral immune response; and main clinical laboratory parameters as indicators for disease severity and mortality in patients with critical and mild COVID-19 without comorbidities or immune-mediated diseases in Saudi Arabia. We determined the expression levels of major proinflammatory cytokines and chemokines; C-reactive protein (CRP); procalcitonin; SARS-CoV-2 IgM antibody and twenty-two clinical laboratory parameters and assessed their usefulness as indicators of disease severity and in-hospital death. Our results showed a significant increase in the expression levels of SARS-CoV-2 IgM antibody; IL1-β; IL-6; IL-8; TNF-α and CRP in critical COVID-19 patients; neutrophil count; urea; creatinine and troponin were also increased. The elevation of these biomarkers was significantly associated and positively correlated with in-hospital death in critical COVID-19 patients. Our results suggest that the levels of IL1-β; IL-6; IL-8; TNF-α; and CRP; neutrophil count; urea; creatinine; and troponin could be used to predict disease severity in COVID-19 patients without comorbidities or immune-mediated diseases. These inflammatory mediators could be used as predictive early biomarkers of COVID-19 disease deterioration; shock and death among COVID-19 patients without comorbidities or immune-mediated diseases.
Collapse
Affiliation(s)
- Muaawia Hamza
- Research Center, King Fahad Medical City, Riyadh, Saudi Arabia.,Faculty of Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Muhanad Alhujaily
- Department of Clinical Laboratory, College of Applied Medicine, University of Bisha, Bisha, Saudi Arabia
| | - Bandar Alosaimi
- Research Center, King Fahad Medical City, Riyadh, Saudi Arabia.,Faculty of Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Karim El Bakkouri
- Department Project Manage; Microbiology Department, Laboratoire national de santé, Dudelange, Luxembourg
| | | | - Mona Alonazi
- Biochemistry Department, College of Science, King Saud University, Saudi Arabia
| | - Mona Awad Alanazi
- Second Health Cluster, Ministry of Healt, Prince Mohammed Bin Abdulaziz Hospital, Riyadh, Saudi Arabia
| | - Basma Abbass
- Department of Biological Sciences, College of Science, University of Jeddah, Saudi Arabia
| | - Abdulsalam Alshehri
- Second Health Cluster, Ministry of Healt, Prince Mohammed Bin Abdulaziz Hospital, Riyadh, Saudi Arabia
| | - Samia T Al-Shouli
- Immunology Unit, Pathology department, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Wael Alturaiki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudi Arabia
| | | |
Collapse
|
45
|
Ma J, Chen X, Wang X, Liang J, Guo L, Su Y, Hao L, Ren C. The accuracy of soluble urokinase-type plasminogen activator receptor for the diagnosis of neonatal sepsis: a meta-analysis. Front Med (Lausanne) 2023; 10:1169114. [PMID: 37181361 PMCID: PMC10174239 DOI: 10.3389/fmed.2023.1169114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 04/04/2023] [Indexed: 05/16/2023] Open
Abstract
Background Neonatal sepsis is one of the major causes of morbidity and mortality in newborns. However, atypical clinical manifestations and symptoms make the early diagnosis of neonatal sepsis a challenge. Relatively high-serum soluble urokinase-type plasminogen activator receptor (suPAR) has been implicated as a diagnostic biomarker for adult sepsis. Therefore, the meta-analysis is intended to explore the diagnostic value of suPAR for neonatal sepsis. Methods The PubMed, Cochrane Library, Embase, Web of Science, China National Knowledge Infrastructure, China Biological Medicine Disk, and Wanfang databases were retrieved from inception to 31 December 2022 to collect diagnostic accuracy studies about suPAR for neonatal sepsis. Two reviewers independently screened the literature, extracted data, and assessed the risk of bias in the included studies using the quality assessment of diagnostic accuracy studies-2 (QUADAS-2) tool. Then, a meta-analysis was performed using Stata 15.0 software. Results A total of six articles involving eight studies were included. The results of the meta-analysis showed that the pooled sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio were 0.89 [95%CI (0.83-0.93)], 0.94 [95%CI (0.77-0.98)], 14 [95%CI (3.5-55.2)], 0.12 [95%CI (0.08-0.18)], and 117 [95%CI (24-567)], respectively. The area under the curve (AUC) of summary receiver operator characteristic (SROC) curves was 0.92 [95%CI (0.90-0.94)]. Sensitivity analysis confirmed the stability of the results, and publication bias was not observed. Fagan's nomogram results demonstrated the clinical availability of the findings. Conclusion Current evidence suggests that suPAR has potential diagnostic value for neonatal sepsis. Owing to the limited quality of the included studies, more high-quality studies are needed to verify the above conclusion.
Collapse
|
46
|
Zhan K, Wang L, Lin H, Fang X, Jia H, Ma X. Novel inflammatory biomarkers in the prognosis of COVID-19. Ther Adv Respir Dis 2023; 17:17534666231199679. [PMID: 37727063 PMCID: PMC10515606 DOI: 10.1177/17534666231199679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 08/18/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND The central role of inflammatory progression in the development of Coronavirus disease 2019 (COVID-19), especially in severe cases, is indisputable. However, the role of some novel inflammatory biomarkers in the prognosis of COVID-19 remains controversial. OBJECTIVE To assess the effect of some novel inflammatory biomarkers in the occurrence and prognosis of COVID-19. METHODS We systematically retrieved the studies related to COVID-19 and the inflammatory biomarkers of interest. The data of each biomarker in different groups were extracted, then were categorized and pooled. The standardized mean difference was chosen as an effect size measure to compare the difference between groups. RESULTS A total of 90 studies with 12,059 participants were included in this study. We found higher levels of endocan, PTX3, suPAR, sRAGE, galectin-3, and monocyte distribution width (MDW) in the COVID-19 positive groups compared to the COVID-19 negative groups. No significant differences for suPAR and galectin-3 were detected between the severe group and mild/moderate group of COVID-19. In addition, the deaths usually had higher levels of PTX3, sCD14-ST, suPAR, and MDW at admission compared to the survivors. Furthermore, patients with higher levels of endocan, galectin-3, sCD14-ST, suPAR, and MDW usually developed poorer comprehensive clinical prognoses. CONCLUSIONS In summary, this meta-analysis provides the most up-to-date and comprehensive evidence for the role of the mentioned novel inflammatory biomarkers in the prognosis of COVID-19, especially in evaluating death and other poor prognoses, with most biomarkers showing a better discriminatory ability.
Collapse
Affiliation(s)
- Kegang Zhan
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
- College of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| | - Luhan Wang
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hao Lin
- West China School of Clinical Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoyu Fang
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hong Jia
- College of Public Health, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiangyu Ma
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
47
|
Nekrasova LA, Shmakova AA, Samokhodskaya LM, Kirillova KI, Stoyanova SS, Mershina EA, Nazarova GB, Rubina KA, Semina EV, Kamalov AA. The Association of PLAUR Genotype and Soluble suPAR Serum Level with COVID-19-Related Lung Damage Severity. Int J Mol Sci 2022; 23:ijms232416210. [PMID: 36555850 PMCID: PMC9785175 DOI: 10.3390/ijms232416210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Uncovering the risk factors for acute respiratory disease coronavirus 2019 (COVID-19) severity may help to provide a valuable tool for early patient stratification and proper treatment implementation, improving the patient outcome and lowering the burden on the healthcare system. Here we report the results of a single-center retrospective cohort study on 151 severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-infected symptomatic hospitalized adult patients. We assessed the association of several blood test measurements, soluble urokinase receptor (uPAR) serum level and specific single nucleotide polymorphisms of ACE (I/D), NOS3 (rs2070744, rs1799983), SERPINE1 (rs1799768), PLAU (rs2227564) and PLAUR (rs344781, rs2302524) genes, with the disease severity classified by the percentage of lung involvement on computerized tomography scans. Our findings reveal that the T/C genotype of PLAUR rs2302524 was independently associated with a less severe lung damage (odds ratio 0.258 [0.071-0.811]). Along with high C-reactive protein, fibrinogen and soluble uPAR serum levels turned out to be independently associated with more severe lung damage in COVID-19 patients. The identified factors may be further employed as predictors of a possibly severe COVID-19 clinical course.
Collapse
Affiliation(s)
- Ludmila A. Nekrasova
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Anna A. Shmakova
- Koltzov Institute of Developmental Biology, 117334 Moscow, Russia
| | - Larisa M. Samokhodskaya
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Karina I. Kirillova
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Simona S. Stoyanova
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Elena A. Mershina
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Galina B. Nazarova
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Kseniya A. Rubina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Ekaterina V. Semina
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
- Institute of Experimental Cardiology, National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
- Correspondence:
| | - Armais A. Kamalov
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| |
Collapse
|
48
|
COMPARATIVE EVALUATION AND PROGNOSTIC UTILITY OF NEURONAL INJURY BIOMARKERS IN COVID-19 PATIENTS: A PROSPECTIVE STUDY. Shock 2022; 58:507-513. [PMID: 36548642 DOI: 10.1097/shk.0000000000002017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
ABSTRACT Background : COVID-19 disease severity markers include mostly molecules related to not only tissue perfusion, inflammation, and thrombosis, but also biomarkers of neural injury. Clinical and basic research has demonstrated that SARS-COV-2 affects the central nervous system. The aims of the present study were to investigate the role of neural injury biomarkers and to compare them with inflammatory markers in their predictive ability of mortality. Methods : We conducted a prospective observational study in critically ill patients with COVID-19 and in a cohort of patients with moderate/severe disease. S100b, neuron-specific enolase (NSE), and inflammatory markers, including soluble urokinase plasminogen activator receptor (suPAR), were measured on intensive care unit or ward admission, respectively. Statistical comparisons between patient groups were performed for all biomarkers under investigation. Correlations between different biomarkers were tested with Spearman correlation coefficient. Receiver operating characteristic curves were plotted using mortality as the classification variable and the biomarker levels on admission as the prognostic variables. Results : A total of 70 patients with COVID-19 were included in the final analysis. Of all studied biomarkers, s100b had the best predictive ability for death in the intensive care unit, with an area under the curve of 0.73 (0.61-0.83), P = 0.0003. S100b levels correlated with NSE, interleukin (IL)-8, and IL-10 (0.27 < rs < 0.37, P < 0.05), and tended to correlate with suPAR ( rs = 0.26, P = 0.05), but not with the vasopressor dose ( P = 0.62). Conclusion : Among the investigated biomarkers, s100b demonstrated the best predictive ability for death in COVID-19 patients. The overall biomarker profile of the patients implies direct involvement of the nervous system by the novel coronavirus.
Collapse
|
49
|
Athale J, Gallagher J, Busch LM. Management of Severe and Critical COVID-19 Infection with Immunotherapies. Infect Dis Clin North Am 2022; 36:761-775. [PMID: 36328635 PMCID: PMC9293954 DOI: 10.1016/j.idc.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Following the reduction in mortality demonstrated by dexamethasone treatment in severe COVID-19, many targeted immunotherapies have been investigated. Thus far, inhibition of IL-6 and JAK pathways have the most robust data and have been granted Emergency Use Authorization for treatment of severe disease. However, it must be noted that critically ill patients comprised a relatively small proportion of most of the trials of COVID-19 therapeutics, despite bearing a disproportionate burden of morbidity and mortality. Furthermore, the rapidity and fluidity with which clinical trials have been conducted in the pandemic setting have contributed to difficulty in extrapolating available trial data to critically ill patients. The exclusion of many patients requiring invasive mechanical ventilation, preponderance of ordinal scale based endpoints, and frequent lack of blinding are particular challenges. More data is needed to identify beneficial treatments in the complex milieu of critical illness from COVID-19 infection.
Collapse
Affiliation(s)
- Janhavi Athale
- Critical Care Medicine Department, Mayo Clinic, Phoenix, AZ, USA
| | - Jolie Gallagher
- Department of Pharmacy, Emory University Hospital, Atlanta, GA, USA
| | - Lindsay M Busch
- Division of Infectious Diseases, Emory University School of Medicine, 101 Woodruff Memorial Building, Suite 2101, Atlanta, GA 30322, USA; Emory Critical Care Center, Atlanta, GA, USA.
| |
Collapse
|
50
|
Su L, Zhang J, Peng Z. The role of kidney injury biomarkers in COVID-19. Ren Fail 2022; 44:1280-1288. [PMID: 35930243 PMCID: PMC9359166 DOI: 10.1080/0886022x.2022.2107544] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease-2019 (COVID-19) outbreak has been declared a global pandemic. COVID-19-associated acute kidney injury (COVID-19 AKI) is related to a high mortality rate and serves as an independent risk factor for hospital death in patients with COVID-19. Early diagnosis would allow for earlier intervention and potentially improve patient outcomes. The goal of early identification of AKI has been the primary impetus for AKI biomarker research, and several kidney injury biomarkers have been demonstrated to be beneficial in predicting COVID-19 AKI as well as disease progression in COVID-19. Furthermore, such data provide valuable insights into the molecular mechanisms underlying this complex and unique disease and serve as a molecular phenotyping tool that could be utilized to direct clinical intervention. This review focuses on a number of kidney injury biomarkers, such as CysC, NAGAL, KIM-1, L-FABP, IL-18, suPAR, and [TIMP-2] • [IGFBP7], which have been widely studied in common clinical settings, such as sepsis, cardiac surgery, and contrast-induced AKI. We explore the role of kidney injury biomarkers in COVID-19 and discuss what remains to be learned.
Collapse
Affiliation(s)
- Lianjiu Su
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jiahao Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Critical Care Medicine, Center of Critical Care Nephrology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|