101
|
Vanassche T, Engelen MM, Orlando C, Vandenbosch K, Gadisseur A, Hermans C, Jochmans K, Minon JM, Motte S, Peperstraete H, Péters P, Sprynger M, Lancellotti P, Dehaene I, Emonts P, Vandenbriele C, Verhamme P, Oury C. The 2023 Belgian clinical guidance on anticoagulation management in hospitalized and ambulatory COVID-19 patients. Acta Clin Belg 2023; 78:497-508. [PMID: 37548503 DOI: 10.1080/17843286.2023.2241692] [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: 02/22/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023]
Abstract
COVID-19 is associated with an increased risk for thrombotic complications. The trials investigating the optimal thromboprophylactic dose are performed in challenging times and seemingly produce conflicting evidence. The burdensome circumstances, divergent endpoints, and different analytical approaches hamper comparison and extrapolation of available evidence. Most importantly, clinicians should provide thromboprophylaxis in hospitalized COVID-19 patients while (re)assessing bleeding and thrombotic risk frequently. The COVID-19 Thromboprophylaxis Working Group of the BSTH updated its guidance document. It aims to summarize the available evidence critically and to guide clinicians in providing the best possible thromboprophylaxis.
Collapse
Affiliation(s)
- Thomas Vanassche
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Matthias M Engelen
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Christelle Orlando
- Department of Haematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Kristel Vandenbosch
- Department of Laboratory Haematology, CHU University Hospital of Liege, Liege, Belgium
| | - Alain Gadisseur
- Department of Haematology, Antwerp University Hospital, Antwerp, Belgium
| | | | - Kristin Jochmans
- Department of Haematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Jean-Marc Minon
- Department of Laboratory Medicine, Thrombosis-Haemostasis and Transfusion Unit, CHR Citadelle, Liege, Belgium
| | - Serge Motte
- Department of Vascular Diseases, Erasme University Hospital, Brussels, Belgium
| | | | - Pierre Péters
- Department of Laboratory Haematology, CHU University Hospital of Liege, Liege, Belgium
| | - Muriel Sprynger
- Department of Cardiology, CHU University Hospital of Liege, Liege, Belgium
| | | | - Isabelle Dehaene
- Vlaamse Vereniging Voor Obstetrie En Gynaecologie, Universitair Ziekenhuis Gent, Ghent, Belgium
| | - Patrick Emonts
- Groupement des Gynecologues Obstetriciens de Langue Francaise de Belgique, CHU University Hospital of Liege, Liège, Belgium
| | | | - Peter Verhamme
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Cecile Oury
- Laboratory of Cardiology, GIGA Institute, University of Liege, Liege, Belgium
| |
Collapse
|
102
|
Guevara S, Miyara SJ, Aronsohn J, Homsi JT, McCann-Molmenti A, Mumford JM, Keber B, Shore-Lesserson L, Morales L, Metz CN, Cho YM, Molmenti CLS, Loto R, Pesce MM, Zafeiropoulos S, Giannis D, Pipolo DO, Jacque F, Montorfano L, Shinozaki K, Shoaib M, Choudhary RC, Nishikimi M, Takegawa R, Endo Y, Hayashida K, Fontan FM, Becker LB, Molmenti EP. COVID-19-Associated Portal Vein Thrombosis Post-Cholecystitis. Int J Angiol 2023; 32:262-268. [PMID: 37927847 PMCID: PMC10624542 DOI: 10.1055/s-0042-1743409] [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: 10/18/2022] Open
Abstract
This case study describes a 45-year-old Caucasian male with a past medical history of obesity, hypertension, and non-insulin-dependent diabetes mellitus, who in the setting of coronavirus disease 2019 (COVID-19) pneumonia, developed portal vein thrombosis (PVT) presenting as an acute abdomen after hospital discharge from a cholecystitis episode. PVT is a very infrequent thromboembolic condition, classically occurring in patients with systemic conditions such as cirrhosis, malignancy, pancreatitis, diverticulitis, autoimmunity, and thrombophilia. PVT can cause serious complications, such as intestinal infarction, or even death, if not promptly treated. Due to the limited number of reports in the literature describing PVT in the COVID-19 setting, its prevalence, natural history, mechanism, and precise clinical features remain unknown. Therefore, clinical suspicion should be high for PVT, in any COVID-19 patient who presents with abdominal pain or associated signs and symptoms. To the best of our knowledge, this is the first report of COVID-19-associated PVT causing extensive thrombosis in the portal vein and its right branch, occurring in the setting of early-stage cirrhosis after a preceding episode of cholecystitis.
Collapse
Affiliation(s)
- Sara Guevara
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Department of Family Medicine, Glen Cove Hospital, Glen Cove, New York
| | - Santiago J. Miyara
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York
| | - Judith Aronsohn
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Department of Anesthesiology, North Shore University Hospital, Manhasset, New York
| | - Joseph T. Homsi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - James M. Mumford
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Department of Family Medicine, Glen Cove Hospital, Glen Cove, New York
| | - Barbara Keber
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Department of Family Medicine, Glen Cove Hospital, Glen Cove, New York
| | - Linda Shore-Lesserson
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Department of Anesthesiology, North Shore University Hospital, Manhasset, New York
| | - Luis Morales
- Department of Surgery, North Shore University Hospital, Manhasset, New York
| | - Christine N. Metz
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York
| | - Young Min Cho
- Department of Internal Medicine, Northeast Georgia Medical Center, Gainesville, Georgia
| | | | - Rodrigo Loto
- Department of Radiology, Delta Sanatorium, Rosario, Santa Fe, ARG
| | - Martin M. Pesce
- Department of Radiology, Delta Sanatorium, Rosario, Santa Fe, ARG
| | - Stefanos Zafeiropoulos
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York
| | | | - Derek O. Pipolo
- Department of Surgery, North Shore University Hospital, Manhasset, New York
| | - Francky Jacque
- Department of Surgery, North Shore University Hospital, Manhasset, New York
| | | | - Koichiro Shinozaki
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Muhammad Shoaib
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Rishabh C. Choudhary
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Mitsuaki Nishikimi
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Ryosuke Takegawa
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Yusuke Endo
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Kei Hayashida
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Fermin M. Fontan
- Department of Surgery, Memorial Medical Center, Las Cruces, New Mexico
| | - Lance B. Becker
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Ernesto P. Molmenti
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Surgery, North Shore University Hospital, Manhasset, New York
| |
Collapse
|
103
|
Landi A, Morici N, Vranckx P, Frigoli E, Bonacchini L, Omazzi B, Tresoldi M, Camponovo C, Moccetti T, Valgimigli M. Edoxaban and/or colchicine in outpatients with COVID-19: rationale and design of the CONVINCE trial. J Cardiovasc Med (Hagerstown) 2023; 24:920-930. [PMID: 37942793 DOI: 10.2459/jcm.0000000000001556] [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: 11/10/2023]
Abstract
BACKGROUND An excessive inflammatory response and a hypercoagulable state are not infrequent in patients with coronavirus disease-2019 (COVID-19) and are associated with adverse clinical outcomes. However, the optimal treatment strategy for COVID-19 patients managed in the out-of-hospital setting is still uncertain. DESIGN The CONVINCE (NCT04516941) is an investigator-initiated, open-label, blinded-endpoint, 2 × 2 factorial design randomized trial aimed at assessing two independently tested hypotheses (anticoagulation and anti-inflammatory ones) in COVID-19 patients. Adult symptomatic patients (≥18 years of age) within 7 days from reverse transcription-PCR (RT-PCR) diagnosis of SARS-CoV-2 infection managed at home or in nursery settings were considered for eligibility. Eligible patients fulfilling all inclusion and no exclusion criteria were randomized to edoxaban versus no treatment (anticoagulation hypothesis) and colchicine versus no treatment (anti-inflammatory hypothesis) in a 1 : 1:1 : 1 ratio. The study had two co-primary endpoints (one for each randomization), including the composite of major vascular thrombotic events at 25 ± 3 days for the anticoagulation hypothesis and the composite of SARS-CoV-2 detection rates at 14 ± 3 days by RT-PCR or freedom from death or hospitalizations (anti-inflammatory hypothesis). Study endpoints will be adjudicated by a blinded Clinical Events Committee. With a final sample size of 420 patients, this study projects an 80% power for each of the two primary endpoints appraised separately. CONCLUSION The CONVINCE trial aims at determining whether targeting anticoagulation and/or anti-inflammatory pathways may confer benefit in COVID-19 patients managed in the out-of-hospital setting. TRIAL REGISTRATION ClinicalTrials.gov number, NCT04516941.
Collapse
Affiliation(s)
- Antonio Landi
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale (EOC)
- Department of Biomedical Sciences, University of Italian Switzerland, Lugano, Switzerland
| | - Nuccia Morici
- IRCCS S. Maria Nascente - Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Pascal Vranckx
- the Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Enrico Frigoli
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale (EOC)
| | - Luca Bonacchini
- Emergency Department, ASST Great Metropolitan Hospital Niguarda, Milan
| | - Barbara Omazzi
- Emergency Unit, ASST Rhodense, Garbagnate Milanese, Italy
| | - Moreno Tresoldi
- Unit of General Medicine and Advanced Care, IRCCS San Raffaele Hospital, Milan
| | - Claudio Camponovo
- Department of Anesthesiology, Clinica Ars Medica, Genolier Swiss Medical Network, Gravesano
| | | | - Marco Valgimigli
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale (EOC)
- Department of Biomedical Sciences, University of Italian Switzerland, Lugano, Switzerland
- University of Bern, Bern, Switzerland
| |
Collapse
|
104
|
de Maistre E, Savard P, Guinot PG. COVID-19 and the Concept of Thrombo-Inflammation: Review of the Relationship between Immune Response, Endothelium and Coagulation. J Clin Med 2023; 12:7245. [PMID: 38068297 PMCID: PMC10706970 DOI: 10.3390/jcm12237245] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 09/11/2024] Open
Abstract
COVID-19, caused by the SARS-CoV-2 virus, has revealed a complex interplay between inflammation and coagulation, leading to the emergence of the concept of thrombo-inflammation. This concept recognizes that COVID-19 is not solely a respiratory illness, but a systemic disease with significant vascular and hematological components. COVID-19 is associated with an unusual prothrombotic state, with intense endothelial activation leading to vasculopathy, cytokine storm, complement system activation and a hypercoagulability state (the activation of platelets and the coagulation cascade, impaired fibrinolysis). The aim of this review is to discuss the different pathological pathways described in COVID-19 that lead to thromboembolic events. Widespread vaccination and post-COVID-19 immunization allows control over the severity of this pandemic. A better understanding of the pathophysiology of COVID-19 can improve the management of frail patients who are hospitalized in intensive care units.
Collapse
Affiliation(s)
| | - Philippe Savard
- Haemostais Unit, Dijon University Hospital, F-21000 Dijon, France;
| | - Pierre-Gregoire Guinot
- Department of Anesthesiology and Intensive Care, Dijon University Hospital, F-21000 Dijon, France;
| |
Collapse
|
105
|
Alizad G, Ayatollahi AA, Shariati Samani A, Samadizadeh S, Aghcheli B, Rajabi A, Nakstad B, Tahamtan A. Hematological and Biochemical Laboratory Parameters in COVID-19 Patients: A Retrospective Modeling Study of Severity and Mortality Predictors. BIOMED RESEARCH INTERNATIONAL 2023; 2023:7753631. [PMID: 38027038 PMCID: PMC10676280 DOI: 10.1155/2023/7753631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/08/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023]
Abstract
Background It is well known that laboratory markers could help in identifying risk factors of severe illness and predicting outcomes of diseases. Here, we performed a retrospective modeling study of severity and mortality predictors of hematological and biochemical laboratory parameters in Iranian COVID-19 patients. Methods Data were obtained retrospectively from medical records of 564 confirmed Iranian COVID-19 cases. According to the disease severity, the patients were categorized into two groups (severe or nonsevere), and based on the outcome of the disease, patients were divided into two groups (recovered or deceased). Demographic and laboratory data were compared between groups, and statistical analyses were performed to define predictors of disease severity and mortality in the patients. Results The study identified a panel of hematological and biochemical markers associated with the severe outcome of COVID-19 and constructed different predictive models for severity and mortality. The disease severity and mortality rate were significantly higher in elderly inpatients, whereas gender was not a determining factor of the clinical outcome. Age-adjusted white blood cells (WBC), platelet cells (PLT), neutrophil-to-lymphocyte ratio (NLR), red blood cells (RBC), hemoglobin (HGB), hematocrit (HCT), erythrocyte sedimentation rate (ESR), mean corpuscular hemoglobin (MCHC), blood urea nitrogen (BUN), and creatinine (Cr) also showed high accuracy in predicting severe cases at the time of hospitalization, and logistic regression analysis suggested grouped hematological parameters (age, WBC, NLR, PLT, HGB, and international normalized ratio (INR)) and biochemical markers (age, BUN, and lactate dehydrogenase (LDH)) as the best models of combined laboratory predictors for severity and mortality. Conclusion The findings suggest that a panel of several routine laboratory parameters recorded on admission could be helpful for clinicians to predict and evaluate the risk of disease severity and mortality in COVID-19 patients.
Collapse
Affiliation(s)
- Ghazaleh Alizad
- Department of Immunology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Asghar Ayatollahi
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Saeed Samadizadeh
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Bahman Aghcheli
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abdolhalim Rajabi
- Environmental Health Research Center, Biostatistics & Epidemiology Department, Faculty of Health, Golestan University of Medical Sciences, Gorgan, Iran
| | - Britt Nakstad
- Division of Paediatric and Adolescent Medicine, University of Oslo, Oslo, Norway
- Department of Paediatrics and Adolescent Health, University of Botswana, Gaborone, Botswana
| | - Alireza Tahamtan
- School of International, Golestan University of Medical Sciences, Gorgan, Iran
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
106
|
Unat ÖS, Karimov Z, Serçe Unat D, Damar G, Çağlayan P, Teymurlu F, Sezai Taşbakan M, Korkmaz Ekren P, Başoğlu Ö, Sayıner A. What is the Optimal Treatment Regimen of Low-MolecularWeight Heparin in Coronavirus Disease 2019 Pneumonia? THORACIC RESEARCH AND PRACTICE 2023; 25:5 - 10. [PMID: 37970682 PMCID: PMC11158007 DOI: 10.5152/thoracrespract.2023.23039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 09/06/2023] [Indexed: 11/17/2023]
Abstract
OBJECTIVE The optimal anticoagulant treatment regimen in hospitalized coronavirus disease 2019 (COVID-19) patients is uncertain. This study aimed to compare the rates of disease progression and mortality in patients treated with low-molecular-weight heparin (LMWH) according to baseline d-dimer levels and in those who received a fixed-dose regimen irrespective of the d-dimer level. MATERIAL AND METHODS This was a retrospective analysis of all patients admitted to a university hospital for COVID-19 pneumonia during a 1-year period. The protocol for d-dimer-driven therapy (on-protocol) was as follows: prophylactic dose when the baseline level is <1000 ng/mL, intermediate dose when the level is between 1000 and 3000 ng/mL, and therapeutic dose when the level is >3000 ng/mL. We compared the progression and mortality rates between the on-protocol and off-protocol treatment groups. The offprotocol group consisted of patients that received a fixed-dose LMWH regimen, which was not in accordance with the defined protocol. RESULTS Of 384 patients (mean age 61.5 ± 15.9 years, 216 male), 294 patients with complete data composed the study group, and 174 patients were treated on-protocol and 120 patients were treated off-protocol. The on-protocol group had lower C-reactive protein (CRP), ferritin, lactate dehydrogenase (LDH), and d-dimer levels and higher SpO2/FiO2 levels at admission. Disease progression developed in 45/174 on-protocol patients (25.9%) vs. 53/120 off-protocol patients (44.2%) during the follow-up (P = .001), and mortality was 29 (16.7%) vs. 32 (26.7%), respectively (P = .041). Logistic regression analysis was performed and included age, presence of comorbidities, LMWH regimen, baseline SpO2/FiO2, CRP, and LDH levels as independent variables. The presence of cardiac comorbidity, age, CRP, and LDH levels, but not the LMWH treatment regimen, were associated with both disease progression and mortality. CONCLUSION A d-dimer-driven LMWH treatment protocol is not associated with better clinical outcomes in hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Ömer Selim Unat
- Department of Pulmonology, Ege University Faculty of Medicine, İzmir, Turkey
| | - Ziya Karimov
- Medicine Program, Ege University Faculty of Medicine, İzmir, Turkey
| | - Damla Serçe Unat
- Department of Pulmonology, İzmir Kemalpaşa State Hospital, İzmir, Turkey
| | - Gizem Damar
- Department of Pulmonology, Ege University Faculty of Medicine, İzmir, Turkey
| | - Pakize Çağlayan
- Department of Pulmonology, Ege University Faculty of Medicine, İzmir, Turkey
| | - Faik Teymurlu
- Department of Pulmonology, Ege University Faculty of Medicine, İzmir, Turkey
| | | | | | - Özen Başoğlu
- Department of Pulmonology, Ege University Faculty of Medicine, İzmir, Turkey
| | - Abdullah Sayıner
- Department of Pulmonology, Ege University Faculty of Medicine, İzmir, Turkey
| |
Collapse
|
107
|
Borulu F, Ceyhun G, Çalik ES, Jalalzai I, Usta H. Multiple Arterial Thrombosis after COVID-19: A Case Report. Vasc Specialist Int 2023; 39:34. [PMID: 37936477 PMCID: PMC10630740 DOI: 10.5758/vsi.230063] [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: 07/03/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 11/09/2023] Open
Abstract
Since the beginning of severe acute respiratory syndrome Coronavirus 2 pandemic, many reports have pointed to states of incrieased hypercoagulability during the acute phase of the disease. We report a 63-year-old female who developed acute mesenteric ischemia due to celiac trunk and superior mesenteric artery thrombi together with acute lower extremity ischemia caused by saddle embolism of the iliac bifurcation and thrombosis of the left external iliac artery. These thrombi developed 20 days after discharge from an intensive care unit due to severe pneumonia and pulmonary embolism associated with COVID-19. The patient had consecutive interventions. Surgical thrombectomy for aortoiliac thrombosis was performed and the mesenteric thrombosis was treated by percutaneous endovascular intervention. We emphasize that the prothrombotic state after COVID-19 infection may persist long after the acute symptomatic phase.
Collapse
Affiliation(s)
- Ferhat Borulu
- Department of Cardiovascular Surgery, Ordu University Medical Faculty, Ordu, Turkey
| | - Gökhan Ceyhun
- Departments of Cardiology, Ataturk University Medical Faculty, Erzurum, Turkey
| | - Eyüp Serhat Çalik
- Departments of Cardiovascular Surgery, Ataturk University Medical Faculty, Erzurum, Turkey
| | - Izatullah Jalalzai
- Departments of Cardiovascular Surgery, Ataturk University Medical Faculty, Erzurum, Turkey
| | - Hakan Usta
- Departments of Cardiovascular Surgery, Ataturk University Medical Faculty, Erzurum, Turkey
| |
Collapse
|
108
|
Möller M, Borg K, Janson C, Lerm M, Normark J, Niward K. Cognitive dysfunction in post-COVID-19 condition: Mechanisms, management, and rehabilitation. J Intern Med 2023; 294:563-581. [PMID: 37766515 DOI: 10.1111/joim.13720] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
The long-term effects of COVID-19 on cognitive function have become an area of increasing concern. This paper provides an overview of characteristics, risk factors, possible mechanisms, and management strategies for cognitive dysfunction in post-COVID-19 condition (PCC). Prolonged cognitive dysfunction is one of the most common impairments in PCC, affecting between 17% and 28% of the individuals more than 12 weeks after the infection and persisting in some cases for several years. Cognitive dysfunctions can be manifested as a wide range of symptoms including memory impairment, attention deficit, executive dysfunction, and reduced processing speed. Risk factors for developing PCC, with or without cognitive impairments, include advanced age, preexisting medical conditions, and the severity of acute illness. The underlying mechanisms remain unclear, but proposed contributors include neuroinflammation, hypoxia, vascular damage, and latent virus reactivation not excluding the possibility of direct viral invasion of the central nervous system, illustrating complex viral pathology. As the individual variation of the cognitive impairments is large, a neuropsychological examination and a person-centered multidimensional approach are required. According to the World Health Organization, limited evidence on COVID-19-related cognitive impairments necessitates implementing rehabilitation interventions from established practices of similar conditions. Psychoeducation and compensatory skills training are recommended. Assistive products and environmental modifications adapted to individual needs might be helpful. In specific attention- and working memory dysfunctions, cognitive training-carefully monitored for intensity-might be effective for people who do not suffer from post-exertional malaise. Further research is crucial for evidence-based interventions specific to COVID-19-related cognitive impairments.
Collapse
Affiliation(s)
- Marika Möller
- Department of Clinical Sciences, Division of Rehabilitation Medicine, Karolinska Institutet, Danderyd University Hospital, Stockholm, Sweden
| | - Kristian Borg
- Department of Clinical Sciences, Division of Rehabilitation Medicine, Karolinska Institutet, Danderyd University Hospital, Stockholm, Sweden
| | - Christer Janson
- Department of Medical Sciences: Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Maria Lerm
- Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection, Linköping University, Linköping, Sweden
| | - Johan Normark
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Katarina Niward
- Department of Infectious Diseases, and Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection, Linköping University, Linköping, Sweden
| |
Collapse
|
109
|
Hibiya S, Fujii T, Fujii T, Suzuki S, Kondo M, Ooka S, Furumoto Y, Azuma S, Tanaka K, Kurata H, Tanaka S, Kurosaki M, Nagayama K, Kusano F, Iizuka Y, Kawamura T, Ikemiyagi H, Sakita S, Yauchi T, Watanabe H, Kawamoto A, Matsuyama Y, Ohtsuka K, Okamoto R. COVID-19 severity is associated with the risk of gastrointestinal bleeding. BMJ Open Gastroenterol 2023; 10:e001199. [PMID: 37963649 PMCID: PMC10649400 DOI: 10.1136/bmjgast-2023-001199] [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: 05/30/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
OBJECTIVE The association between the severity of COVID-19 and gastrointestinal (GI) bleeding is unknown. This study aimed to determine whether the severity of COVID-19 is a risk factor for GI bleeding. DESIGN A multicentre, retrospective cohort study was conducted on hospitalised patients with COVID-19 between January 2020 and December 2021. The severity of COVID-19 was classified according to the National Institute of Health severity classification. The primary outcome was the occurrence of GI bleeding during hospitalisation. The main analysis compared the relationship between the severity of COVID-19 and the occurrence of GI bleeding. Multivariable logistic regression analysis was performed to evaluate the association between the severity of COVID-19 and the occurrence of GI bleeding. RESULTS 12 044 patients were included. 4165 (34.6%) and 1257 (10.4%) patients had severe and critical COVID-19, respectively, and 55 (0.5%) experienced GI bleeding. Multivariable analysis showed that patients with severe COVID-19 had a significantly higher risk of GI bleeding than patients with non-severe COVID-19 (OR: 3.013, 95% CI: 1.222 to 7.427). Patients with critical COVID-19 also had a significantly higher risk of GI bleeding (OR: 15.632, 95% CI: 6.581 to 37.130). Patients with severe COVID-19 had a significantly increased risk of lower GI bleeding (OR: 10.349, 95% CI: 1.253 to 85.463), but the risk of upper GI bleeding was unchanged (OR: 1.875, 95% CI: 0.658 to 5.342). CONCLUSION The severity of COVID-19 is associated with GI bleeding, and especially lower GI bleeding was associated with the severity of COVID-19. Patients with severe or critical COVID-19 should be treated with caution as they are at higher risk for GI bleeding.
Collapse
Affiliation(s)
- Shuji Hibiya
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takashi Fujii
- Department of Gastroenterology, Tokyo Metropolitan Hiroo Hospital, Tokyo, Japan
| | - Toshimitsu Fujii
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Suzuki
- Department of Gastroenterology, Tokyo Metropolitan Hiroo Hospital, Tokyo, Japan
| | - Mayumi Kondo
- Department of Gastroenterology, Tokyo Metropolitan Toshima Hospital, Tokyo, Japan
| | - Shinya Ooka
- Department of Gastroenterology, Tokyo Metropolitan Toshima Hospital, Tokyo, Japan
| | - Yohei Furumoto
- Department of Gastroenterology, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
| | - Seishin Azuma
- Department of Gastroenterology, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
| | - Kei Tanaka
- Department of Gastroenterology, Tokyo Metropolitan Otsuka Hospital, Tokyo, Japan
| | - Hitoshi Kurata
- Department of Gastroenterology, Tokyo Metropolitan Otsuka Hospital, Tokyo, Japan
| | - Shohei Tanaka
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Masayuki Kurosaki
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| | | | - Fumihiko Kusano
- Department of Gastroenterology, Tsuchiura Kyodo General Hospital, Ibaraki, Japan
| | - Yasuhiro Iizuka
- Department of Gastroenterology, Kashiwa Municipal Hospital, Chiba, Japan
| | - Takahiro Kawamura
- Department of Gastroenterology, JA Toride Medical Center, Ibaraki, Japan
| | - Hidekazu Ikemiyagi
- Department of Gastroenterology, Yokohama City Minato Red Cross Hospital, Kanagawa, Japan
| | - Shinya Sakita
- Department of Gastroenterology, Yokohama City Minato Red Cross Hospital, Kanagawa, Japan
| | - Tsunehito Yauchi
- Department of Gastroenterology, Soka Municipal Hospital, Saitama, Japan
| | - Hideki Watanabe
- Department of Gastroenterology, Yokosuka Kyosai Hospital, Kanagawa, Japan
| | - Ami Kawamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Matsuyama
- Department of Oral Health Promotion, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuo Ohtsuka
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
110
|
Escobar C, Bover Freire R, García-Moll Marimón X, González-Juanatey C, Morillas M, Valle Muñoz A, Gómez Doblas JJ. A Delphi consensus on the management of anticoagulation in the COVID-19 pandemic: the MONACO study. Cardiovasc Diagn Ther 2023; 13:777-791. [PMID: 37941839 PMCID: PMC10628427 DOI: 10.21037/cdt-23-76] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 09/15/2023] [Indexed: 11/10/2023]
Abstract
Background During the COVID-19 pandemic, guideline documents on the management of anticoagulation were rapidly published. However, these documents did not follow a structured methodology, and significant differences existed between the guidelines. The aim of this expert consensus was to provide recommendations on the clinical management of oral anticoagulation in patients in the context of the COVID-19 pandemic. Methods A two-round Delphi study was conducted using an online survey. In the first round, panellists expressed their level of agreement with the items on a 9-point Likert scale. Items were selected if they received approval from ≥66.6% of panellists and if they were agreed by the scientific committee. In the second round, panellists revaluated those items that did not meet consensus in the first round. Results A total of 147 panellists completed the first round, and 144 of them completed the second round. Consensus was reached on 161 items included in five dimensions. These dimensions addressed: (I) management of anticoagulation in patients with atrial fibrillation (AF) without mechanical valves or moderate/severe mitral stenosis during COVID-19 infection; (II) thromboprophylaxis in patients hospitalised for COVID-19; (III) management of anticoagulation at hospital discharge/after COVID-19; (IV) anticoagulation monitoring in the COVID-19 pandemic setting; and (V) role of telemedicine in the management and follow-up of patients with AF in the COVID-19 pandemic setting. Conclusions These areas of collective agreement could specially guide clinicians in making decisions regarding anticoagulation in patients with COVID-19 during hospitalisation and at discharge, where results from clinical trials are still limited and, in some cases, conflicting.
Collapse
Affiliation(s)
- Carlos Escobar
- Cardiology Department, Hospital Universitario La Paz, Madrid, Spain
| | | | | | | | - Miren Morillas
- Cardiology Department, Hospital de Galdakao, Galdakao, Spain
| | | | - Juan José Gómez Doblas
- Cardiology Department, Hospital Universitario Virgen de la Victoria, CIBERCV, Málaga, Spain
| |
Collapse
|
111
|
Casillas N, Ramón A, Torres AM, Blasco P, Mateo J. Predictive Model for Mortality in Severe COVID-19 Patients across the Six Pandemic Waves. Viruses 2023; 15:2184. [PMID: 38005862 PMCID: PMC10675561 DOI: 10.3390/v15112184] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/21/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
The impact of SARS-CoV-2 infection remains substantial on a global scale, despite widespread vaccination efforts, early therapeutic interventions, and an enhanced understanding of the disease's underlying mechanisms. At the same time, a significant number of patients continue to develop severe COVID-19, necessitating admission to intensive care units (ICUs). This study aimed to provide evidence concerning the most influential predictors of mortality among critically ill patients with severe COVID-19, employing machine learning (ML) techniques. To accomplish this, we conducted a retrospective multicenter investigation involving 684 patients with severe COVID-19, spanning from 1 June 2020 to 31 March 2023, wherein we scrutinized sociodemographic, clinical, and analytical data. These data were extracted from electronic health records. Out of the six supervised ML methods scrutinized, the extreme gradient boosting (XGB) method exhibited the highest balanced accuracy at 96.61%. The variables that exerted the greatest influence on mortality prediction encompassed ferritin, fibrinogen, D-dimer, platelet count, C-reactive protein (CRP), prothrombin time (PT), invasive mechanical ventilation (IMV), PaFi (PaO2/FiO2), lactate dehydrogenase (LDH), lymphocyte levels, activated partial thromboplastin time (aPTT), body mass index (BMI), creatinine, and age. These findings underscore XGB as a robust candidate for accurately classifying patients with COVID-19.
Collapse
Affiliation(s)
- Nazaret Casillas
- Department of Internal Medicine, Hospital Virgen De La Luz, 16002 Cuenca, Spain
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
| | - Antonio Ramón
- Department of Pharmacy, General University Hospital, 46014 Valencia, Spain
| | - Ana María Torres
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Pilar Blasco
- Department of Pharmacy, General University Hospital, 46014 Valencia, Spain
| | - Jorge Mateo
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| |
Collapse
|
112
|
Abdulsalam H, Li J, Loka RS, Sletten ET, Nguyen HM. Heparan Sulfate-Mimicking Glycopolymers Bind SARS-CoV-2 Spike Protein in a Length- and Sulfation Pattern-Dependent Manner. ACS Med Chem Lett 2023; 14:1411-1418. [PMID: 37849547 PMCID: PMC10577887 DOI: 10.1021/acsmedchemlett.3c00319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/27/2023] [Indexed: 10/19/2023] Open
Abstract
Heparan sulfate-mimicking glycopolymers, composed of glucosamine (GlcN)-glucuronic acid (GlcA) repeating units, bind to the receptor-binding subunit (S1) and spike glycoprotein (S) domains of the SARS-CoV-2 spike protein in a length- and sulfation pattern-dependent fashion. A glycopolymer composed of 12 repeating GlcNS6S-GlcA units exhibits a much higher affinity to the S1 protein (IC50 = 13 ± 1.1 nM) compared with the receptor-binding domain (RBD). This glycopolymer does not interfere in angiotensin-converting enzyme 2 binding of the RBD. Although this compound binds strongly to the S1/membrane-fusion subunit (S2) junction (KD = 29.7 ± 4.18 nM), it does not shield the S1/S2 site from cleavage by furin-a behavior contrary to natural heparin. This glycopolymer lacks iduronic acid, which accounts for 70% of heparin. Further, this compound, unlike natural heparin, is well defined in both sulfation pattern and length, which results in fewer off-target interactions with heparin-binding proteins. The results highlight the potential of using polymeric heparan sulfate (HS) mimetics for the therapeutic agent development.
Collapse
Affiliation(s)
- Hawau Abdulsalam
- Department
of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Jiayi Li
- Department
of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Ravi S. Loka
- Department
of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Eric T. Sletten
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, Am Muehlenberg 1, 14476 Potsdam, Germany
| | - Hien M. Nguyen
- Department
of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| |
Collapse
|
113
|
Gałgańska H, Jarmuszkiewicz W, Gałgański Ł. Carbon dioxide and MAPK signalling: towards therapy for inflammation. Cell Commun Signal 2023; 21:280. [PMID: 37817178 PMCID: PMC10566067 DOI: 10.1186/s12964-023-01306-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/05/2023] [Indexed: 10/12/2023] Open
Abstract
Inflammation, although necessary to fight infections, becomes a threat when it exceeds the capability of the immune system to control it. In addition, inflammation is a cause and/or symptom of many different disorders, including metabolic, neurodegenerative, autoimmune and cardiovascular diseases. Comorbidities and advanced age are typical predictors of more severe cases of seasonal viral infection, with COVID-19 a clear example. The primary importance of mitogen-activated protein kinases (MAPKs) in the course of COVID-19 is evident in the mechanisms by which cells are infected with SARS-CoV-2; the cytokine storm that profoundly worsens a patient's condition; the pathogenesis of diseases, such as diabetes, obesity, and hypertension, that contribute to a worsened prognosis; and post-COVID-19 complications, such as brain fog and thrombosis. An increasing number of reports have revealed that MAPKs are regulated by carbon dioxide (CO2); hence, we reviewed the literature to identify associations between CO2 and MAPKs and possible therapeutic benefits resulting from the elevation of CO2 levels. CO2 regulates key processes leading to and resulting from inflammation, and the therapeutic effects of CO2 (or bicarbonate, HCO3-) have been documented in all of the abovementioned comorbidities and complications of COVID-19 in which MAPKs play roles. The overlapping MAPK and CO2 signalling pathways in the contexts of allergy, apoptosis and cell survival, pulmonary oedema (alveolar fluid resorption), and mechanical ventilation-induced responses in lungs and related to mitochondria are also discussed. Video Abstract.
Collapse
Affiliation(s)
- Hanna Gałgańska
- Faculty of Biology, Molecular Biology Techniques Laboratory, Adam Mickiewicz University in Poznan, Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland
| | - Wieslawa Jarmuszkiewicz
- Faculty of Biology, Department of Bioenergetics, Adam Mickiewicz University in Poznan, Institute of Molecular Biology and Biotechnology, Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland
| | - Łukasz Gałgański
- Faculty of Biology, Department of Bioenergetics, Adam Mickiewicz University in Poznan, Institute of Molecular Biology and Biotechnology, Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland.
| |
Collapse
|
114
|
Kawakami E, Saiki N, Yoneyama Y, Moriya C, Maezawa M, Kawamura S, Kinebuchi A, Kono T, Funata M, Sakoda A, Kondo S, Ebihara T, Matsumoto H, Togami Y, Ogura H, Sugihara F, Okuzaki D, Kojima T, Deguchi S, Vallee S, McQuade S, Islam R, Natarajan M, Ishigaki H, Nakayama M, Nguyen CT, Kitagawa Y, Wu Y, Mori K, Hishiki T, Takasaki T, Itoh Y, Takayama K, Nio Y, Takebe T. Complement factor D targeting protects endotheliopathy in organoid and monkey models of COVID-19. Cell Stem Cell 2023; 30:1315-1330.e10. [PMID: 37802037 PMCID: PMC10575686 DOI: 10.1016/j.stem.2023.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 07/04/2023] [Accepted: 09/01/2023] [Indexed: 10/08/2023]
Abstract
COVID-19 is linked to endotheliopathy and coagulopathy, which can result in multi-organ failure. The mechanisms causing endothelial damage due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain elusive. Here, we developed an infection-competent human vascular organoid from pluripotent stem cells for modeling endotheliopathy. Longitudinal serum proteome analysis identified aberrant complement signature in critically ill patients driven by the amplification cycle regulated by complement factor B and D (CFD). This deviant complement pattern initiates endothelial damage, neutrophil activation, and thrombosis specific to organoid-derived human blood vessels, as verified through intravital imaging. We examined a new long-acting, pH-sensitive (acid-switched) antibody targeting CFD. In both human and macaque COVID-19 models, this long-acting anti-CFD monoclonal antibody mitigated abnormal complement activation, protected endothelial cells, and curtailed the innate immune response post-viral exposure. Collectively, our findings suggest that the complement alternative pathway exacerbates endothelial injury and inflammation. This underscores the potential of CFD-targeted therapeutics against severe viral-induced inflammathrombotic outcomes.
Collapse
Affiliation(s)
- Eri Kawakami
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Norikazu Saiki
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Yosuke Yoneyama
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Chiharu Moriya
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Mari Maezawa
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Shuntaro Kawamura
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Akiko Kinebuchi
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Tamaki Kono
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Masaaki Funata
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Ayaka Sakoda
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Shigeru Kondo
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Takeshi Ebihara
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hisatake Matsumoto
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Yuki Togami
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Fuminori Sugihara
- Core Instrumentation Facility, Immunology Frontier Research Center and Research Institute for Microbial Diseases, Osaka University, 3-3-1, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Disease, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Takashi Kojima
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Sayaka Deguchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Sebastien Vallee
- Rare Disease DDU, Takeda Pharmaceutical Company Ltd, 125 Binney Street, Cambridge, MA 02139, USA
| | - Susan McQuade
- Rare Disease DDU, Takeda Pharmaceutical Company Ltd, 125 Binney Street, Cambridge, MA 02139, USA; BPS Biosciences Inc., 6405 Mira Mesa Blvd. Suite 100, San Diego, CA 92121, USA
| | - Rizwana Islam
- Rare Disease DDU, Takeda Pharmaceutical Company Ltd, 125 Binney Street, Cambridge, MA 02139, USA
| | - Madhusudan Natarajan
- Rare Disease DDU, Takeda Pharmaceutical Company Ltd, 125 Binney Street, Cambridge, MA 02139, USA
| | - Hirohito Ishigaki
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Misako Nakayama
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Cong Thanh Nguyen
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Yoshinori Kitagawa
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Yunheng Wu
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Kensaku Mori
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Information Technology Center, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Research Center for Medical Bigdata, National Institute of Informatics, Tokyo 100-0003, Japan
| | - Takayuki Hishiki
- Kanagawa Prefectural Institute of Public Health, 1-3-1, Shimomachiya, Chigasaki, Kanagawa 253-0087, Japan; Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Tomohiko Takasaki
- Kanagawa Prefectural Institute of Public Health, 1-3-1, Shimomachiya, Chigasaki, Kanagawa 253-0087, Japan; Advanced Technology and Development Division, BML, INC, 1361-1, Matoba, Kawagoe-shi, Saitama 350-1101, Japan
| | - Yasushi Itoh
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Yasunori Nio
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan.
| | - Takanori Takebe
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Division of Gastroenterology, Hepatology and Nutrition & Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; The Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Communication Design Center, Advanced Medical Research Center, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe) and Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
115
|
Mv P, Auanassova A, Yessirkepov M, Zimba O, Gasparyan AY, Kitas GD, Ahmed S. New-onset systemic vasculitis following SARS-CoV-2 infection and vaccination: the trigger, phenotype, and outcome. Clin Rheumatol 2023; 42:2761-2775. [PMID: 37422611 DOI: 10.1007/s10067-023-06694-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/13/2023] [Accepted: 07/02/2023] [Indexed: 07/10/2023]
Abstract
The global health crisis caused by the COVID-19 pandemic overwhelmed the capacity of healthcare systems to cope with the rapidly spreading infection and its associated complications. Among these complications, autoimmune phenomena such as systemic vasculitis emerged as a significant challenge. Both the SARS-CoV-2 virus and the vaccines developed to combat it appeared to induce clinical manifestations resembling various types of systemic vasculitis, affecting large, medium, and small vessels. These virus- or vaccine-induced vasculitides exhibited a distinct natural history and course from de novo vasculitis, as they were more responsive to steroid therapy and some mild cases even resolved spontaneously. Notably, there have been no confirmed cases of SARS-CoV-2 infection or vaccination triggering variable vessel vasculitis like Behcet's disease or Kawasaki disease. IgA vasculitis, which is predominantly a pediatric condition, was more prevalent in adults after COVID-19 infection and they had a favorable outcome with glucocorticoid treatment. The impact of immunosuppression, especially B-cell-depleting agents, on the immunogenicity of the vaccine was evident, but there was no significant increase in the incidence of SARS-CoV-2 infection in these patients compared to the general population. Considering their relatively benign course, these post-COVID or post-vaccine vasculitides seem to be amenable to 0.8 to 1 mg/kg prednisolone or equivalent, which could be gradually tapered. The need for immunosuppression and the duration of steroid therapy should be determined on an individual basis. While the world still reels from the perils of a deadly pandemic, the aftermath continues to haunt. Our narrative review aims to explore the effects of COVID and the vaccine on systemic vasculitis, as well as the effect of disease and immunosuppression on the immunogenicity of the COVID vaccine.
Collapse
Affiliation(s)
- Prakashini Mv
- Department of Clinical Immunology and Rheumatology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, India, 751024
| | - Akerke Auanassova
- Department of Biology and Biochemistry, South Kazakhstan Medical Academy, Shymkent, Kazakhstan
| | - Marlen Yessirkepov
- Department of Biology and Biochemistry, South Kazakhstan Medical Academy, Shymkent, Kazakhstan
| | - Olena Zimba
- Department of Clinical Rheumatology and Immunology, University Hospital in Krakow, Krakow, Poland
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
- Department of Internal Medicine N2, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Armen Yuri Gasparyan
- Departments of Rheumatology and Research and Development, Dudley Group NHS Foundation Trust (Teaching Trust of the University of Birmingham, UK), Russells Hall Hospital, Dudley, West Midlands, UK
| | - George D Kitas
- Departments of Rheumatology and Research and Development, Dudley Group NHS Foundation Trust (Teaching Trust of the University of Birmingham, UK), Russells Hall Hospital, Dudley, West Midlands, UK
- Centre for Epidemiology Versus Arthritis, University of Manchester, Manchester, UK
| | - Sakir Ahmed
- Department of Clinical Immunology and Rheumatology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, India, 751024.
| |
Collapse
|
116
|
Feler J, Chuck C, Anderson M, Poggi J, Sweeney J, Moldovan K, Jayaraman MV, McTaggart R, Torabi R. Dual antiplatelet use in the management of COVID-19 associated acute ischemic stroke reocclusion. Interv Neuroradiol 2023; 29:540-547. [PMID: 35549746 PMCID: PMC10549714 DOI: 10.1177/15910199221097484] [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: 02/25/2022] [Accepted: 04/11/2022] [Indexed: 11/17/2022] Open
Abstract
INTRO SARS-CoV-2 (COVID-19) infection is associated with acute ischemic stroke (AIS), which may be due to a prothrombotic state. Early reports have suggested high rates of reocclusion following mechanical thrombectomy (MT) with poor radiographic and clinical outcomes. We report our early experience using intra-procedural antithrombotics to address SARS-CoV-2 reocclusion. METHODS We identified 6 patients that experienced early reocclusion after MT for COVID-19-associated AIS through retrospective chart review abstracting their basic demographics, COVID-19 status, and stroke management. All these patients were treated after reocclusion with aspirin and cangrelor intra-procedurally, the latter of which was converted to ticagrelor post-procedurally. Some patients additionally received argatroban infusion intraprocedurally. RESULTS Mean age was 54. There were 3 post-procedural and 3 intra-procedural re-occlusions. After repeat thrombectomy and treatment with aspirin and cangrelor, those with post-procedure reocclusion did not show further reocclusion, while those with intra-procedural reocclusion showed radiographic improvement with intraprocedural cangrelor administration. Outcomes for these patients were poor, with a median mRS of 4. Two patients developed petechial hemorrhage of their stroke which was managed conservatively, and one developed a retroperitoneal hemorrhage from femoral access requiring transfusion. There were no patients who developed new parenchymal hematomas. CONCLUSION COVID-19 AIS may be associated with a hypercoagulable state which risks malignant reocclusion complicating MT. We found antithrombotic treatment periprocedural cangrelor with or without argatroban transitioned to oral aspirin with ticagrelor to be a viable method for management of these patients.
Collapse
Affiliation(s)
- Joshua Feler
- Department of Neurosurgery, Brown University, The Warren Alpert Medical School, Providence, RI, USA
| | - Carlin Chuck
- Department of Neurosurgery, Brown University, The Warren Alpert Medical School, Providence, RI, USA
| | - Matthew Anderson
- Department of Neurosurgery, Brown University, The Warren Alpert Medical School, Providence, RI, USA
| | - Jonathan Poggi
- Department of Neurosurgery, Brown University, The Warren Alpert Medical School, Providence, RI, USA
| | - Joseph Sweeney
- Department of Hematology-Oncology, Brown University, The Warren Alpert Medical School, Providence, RI, USA
| | - Krisztina Moldovan
- Department of Interventional Radiology, Brown University, The Warren Alpert Medical School, Providence, RI, USA
| | - Mahesh V. Jayaraman
- Department of Interventional Radiology, Brown University, The Warren Alpert Medical School, Providence, RI, USA
| | - Ryan McTaggart
- Department of Interventional Radiology, Brown University, The Warren Alpert Medical School, Providence, RI, USA
| | - Radmehr Torabi
- Department of Neurosurgery, Brown University, The Warren Alpert Medical School, Providence, RI, USA
| |
Collapse
|
117
|
Pinosanu EA, Burada E, Pirscoveanu D, Aldea M, Albu CV, Surugiu R, Sandu RE. Predictive Value of Pulmonary Involvement in Stroke Patients Co-Infected with COVID-19. CURRENT HEALTH SCIENCES JOURNAL 2023; 49:536-545. [PMID: 38559828 PMCID: PMC10976201 DOI: 10.12865/chsj.49.04.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/12/2023] [Indexed: 04/04/2024]
Abstract
In response to the intricate clinical challenges posed by the intersection of COVID-19 and acute ischemic stroke, the Neuropsychiatry Hospital of Craiova, Romania, initiated a comprehensive study. This research aims to unravel the impacts of pulmonary complications on ischemic stroke outcomes, comparing patients with concurrent SARS-CoV-2 infection to those without. The study integrates pulmonary assessments, acknowledging the significant role respiratory involvement plays in the progression and prognosis of stroke patients during the pandemic. By systematically examining individuals with both acute ischemic stroke and COVID-19, the study seeks to shed light on the complex interplay between cerebral and pulmonary health. The findings are expected to enhance patient care by informing clinical decisions and leading to more effective management approaches for stroke patients in the COVID-19 era.
Collapse
Affiliation(s)
- Elena Anca Pinosanu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, Romania
| | - Emilia Burada
- Department of Physiology, University of Medicine and Pharmacy of Craiova, Romania
| | - Denisa Pirscoveanu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, Romania
| | - Madalina Aldea
- Department of Psychiatry, University of Medicine and Pharmacy of Craiova, Romania
| | - Carmen Valeria Albu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, Romania
| | - Roxana Surugiu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, Romania
| | - Raluca Elena Sandu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, Romania
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, Romania
| |
Collapse
|
118
|
Baptista A, Vieira AM, Capela E, Julião P, Macedo A. COVID-19 fatality rates in hospitalized patients: A new systematic review and meta-analysis. J Infect Public Health 2023; 16:1606-1612. [PMID: 37579698 DOI: 10.1016/j.jiph.2023.07.006] [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: 03/22/2023] [Revised: 06/21/2023] [Accepted: 07/14/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND SARS-COV2 or COVID-19 disease is an infectious illness that emerged for the first time at the end of 2019, in Wuhan, China and rapidly turned out to be an international pandemic with deleterious effects all over the world. In March 2021, A. Macedo et al., has published the first meta-analysis of hospital mortality, so the authors decided to update those data at a time of emergence of new therapies and increasing vaccination rates. METHODS As the outcome of interest was the mortality in hospitalized general patients, the authors looked for articles evaluating the clinical characteristics of those patients, consulting PUBMED (The US National Library of Medicine) and EMBASE (Medical database) in an independent selection using predefined terms of search. A meta-analysis random-effect model was estimated using Mantel-Haenszel method. Heterogeneity among studies was tested using Tau2 statistics and Chi2 statistics. RESULTS In a first instance 25 articles were included for final analysis with a total of 103,840 patients, but as the goal was to update the anterior data, these studies were analysed together with the 21 studies of the previous meta-analysis, with a total of 114609 patients. The mortality rate of COVID-19 general patients admitted to the hospital was 16% (95% CI 12; 21, I2 =100%). CONCLUSION Global hospital mortality of COVID-19 of general patients was 16%, with quite different rates according to the different geographic areas analysed.
Collapse
Affiliation(s)
- Alexandre Baptista
- Faculdade de Medicina e Ciências Biomédicas Universidade Algarve, Faro, Portugal
| | - Ana M Vieira
- Faculdade de Medicina e Ciências Biomédicas Universidade Algarve, Faro, Portugal
| | - Eunice Capela
- Faculdade de Medicina e Ciências Biomédicas Universidade Algarve, Faro, Portugal
| | - Pedro Julião
- Faculdade de Medicina e Ciências Biomédicas Universidade Algarve, Faro, Portugal
| | - Ana Macedo
- Faculdade de Medicina e Ciências Biomédicas Universidade Algarve, Faro, Portugal; Algarve Biomedical Center, Faro, Portugal.
| |
Collapse
|
119
|
Hao W, Liu M, Bai C, Liu X, Niu S, Chen X. Increased inflammatory mediators levels are associated with clinical outcomes and prolonged illness in severe COVID-19 patients. Int Immunopharmacol 2023; 123:110762. [PMID: 37562295 DOI: 10.1016/j.intimp.2023.110762] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023]
Abstract
OBJECTIVE The purpose of this study was to identify potential predictors of clinical outcome in severe COVID-19 patients and to investigate the relationship between immunological parameters and duration of illness. METHODS This single-center study retrospectively recruited 73 patients with severe or critical COVID-19. Immunological indicators include white blood cell count, neutrophil count, lymphocyte count, neutrophil-to-lymphocyte ratio, and circulating inflammatory mediators were observed for their association with disease severity, mortality and duration of illness of COVID-19. RESULTS Serum inflammatory mediators levels of C-reactive protein (P = 0.015), interleukin 6 (IL-6) (P < 0.001), CX3CL1 (P < 0.001), D-dimer (P < 0.001) and procalcitonin (PCT) (P < 0.001) were increased in critical illness patients compared to those severe COVID-19 patients. CX3CL1 has the highest C-index (0.75) to predict in-hospital mortality in patients with COVID-19. Furthermore, this study shows for the first time that the duration of illness in severe COVID-19 patients is associated with serum levels of CX3CL1 (P = 0.037) and D-dimer (P = 0.014). CONCLUSION CX3CL1, D-dimer, PCT, and IL-6 could effectively predict mortality in severe COVID-19 patients. In addition, only the circulating levels of CX3CL1 and D-dimer were significantly associated with duration of illness.
Collapse
Affiliation(s)
- Wendong Hao
- Department of Allergy, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China; Department of Respiratory and Critical Care Medicine, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China.
| | - Meimei Liu
- Department of Allergy, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China; Department of Respiratory and Critical Care Medicine, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China
| | - Cairong Bai
- Department of Allergy, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China
| | - Xin Liu
- Department of Allergy, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China; Department of Respiratory and Critical Care Medicine, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China
| | - Siqian Niu
- Department of Respiratory and Critical Care Medicine, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China
| | - Xiushan Chen
- Department of Respiratory and Critical Care Medicine, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China
| |
Collapse
|
120
|
Turan Civraz AZ, Duzyol I, Atli E, Caglayan C, Ozer Yurt E, Ata A, Yilmaz M, Karakoyun B. Incidence of Thromboembolism in COVID-19 Patients in Intensive Care Units: A Retrospective Cohort Analysis. Cureus 2023; 15:e47014. [PMID: 37965400 PMCID: PMC10641796 DOI: 10.7759/cureus.47014] [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: 10/13/2023] [Indexed: 11/16/2023] Open
Abstract
INTRODUCTION Coronavirus disease 2019 (COVID-19) infection was declared a pandemic, causing high mortality and morbidity worldwide. It predisposes patients to both arterial and venous thromboembolism, which causes high mortality, and is one of the most serious complications of the disease. OBJECTIVE The aim of this retrospective study was to determine the frequency of thromboembolic events in patients diagnosed with COVID-19 in the intensive care unit (ICU) and to identify the factors causing thromboembolism. MATERIAL AND METHODS The digital records of patients admitted to the adult ICU of Derince Training and Research Hospital, Kocaeli, Turkey, with a diagnosis of COVID-19 between March 13, 2020, and December 31, 2021, were retrospectively reviewed. RESULTS Data of 484 patients, 248 (51.2%) female and 236 (48.8%) male, aged between 18-98 years were analyzed. The overall, arterial and venous incidence of thromboembolism was 14.8%, 11.3%, and 3.5%, respectively. There was no significant association between COVID-19 variants and the development of thromboembolism. The effect of various patient variables on the development of thromboembolism was evaluated, including cardiovascular disease (p<0.001), age (p=0.003), use of acetylsalicylic acid (ASA) (p<0.001), antiplatelet therapy (p<0. 001), acute physiology and chronic health evaluation (APACHE) II score (p=0.003), D-dimer (p=0.015), fibrinogen (p=0.032), ferritin (p=0.015), prothrombin time (PT) (p=0.015), international normalized ratio (INR) (p=0.012), troponin (p=0.012) values at the ICU admission were found statistically significant. The cut-off values were 2.565 (μg/mL) for D-dimer, 435.51 (mg) for fibrinogen, 633.55 (ml/ng) for ferritin, 1.155 for INR, and 0.085 (ng/mL) for troponin. CONCLUSION Although low-molecular-weight heparin (LMWH) is the first choice, it may be appropriate to add ASA and other antiplatelet agents to reduce the risk of thromboembolism in patients with high thromboembolic risk including advanced age, cardiovascular disease, and elevated levels of D-dimer, troponin, ferritin, and fibrinogen.
Collapse
Affiliation(s)
- Ayse Z Turan Civraz
- Department of Anesthesiology and Reanimation, Kocaeli City Hospital, Kocaeli, TUR
| | - Ipek Duzyol
- Department of Anesthesiology and Reanimation, Kocaeli City Hospital, Kocaeli, TUR
| | - Emine Atli
- Department of Anesthesiology and Reanimation, Kocaeli City Hospital, Kocaeli, TUR
| | - Cigdem Caglayan
- Department of Anesthesiology and Reanimation, Kocaeli City Hospital, Kocaeli, TUR
| | - Emine Ozer Yurt
- Department of Anesthesiology and Reanimation, Kocaeli City Hospital, Kocaeli, TUR
| | - Adnan Ata
- Department of Anesthesiology and Reanimation, Kocaeli City Hospital, Kocaeli, TUR
| | - Mehmet Yilmaz
- Department of Anesthesiology and Reanimation, Kocaeli City Hospital, Kocaeli, TUR
| | - Berna Karakoyun
- Department of Physiology, University of Health Sciences, Hamidiye Faculty of Medicine, Istanbul, TUR
| |
Collapse
|
121
|
Ahmadi AS, Shafiei‐Jandaghi NZ, Sadeghi K, Salimi V, Nejati A, Azad TM, Yavarian J. SARS-CoV-2 in patient with protein C deficiency: A case report. Clin Case Rep 2023; 11:e8030. [PMID: 37850062 PMCID: PMC10577157 DOI: 10.1002/ccr3.8030] [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: 06/13/2023] [Revised: 09/18/2023] [Accepted: 09/27/2023] [Indexed: 10/19/2023] Open
Abstract
In SARS-CoV-2 pandemic different disorders in coagulation pathways in COVID-19 patients were reported. We described a 44-year-old female with COVID-19 and protein C deficiency history. She did not show any coagulation disorder during her disease course. Complete genome sequencing of SARS-CoV-2 was performed and some mutations identified and compared with Wuhan strain. Besides hospitalized patients, in COVID-19 outpatients with low concentration of protein C, early prescription of an anticoagulant such as heparin could be helpful in prevention of venous thromboembolism or pulmonary embolism.
Collapse
Affiliation(s)
- Akram Sadat Ahmadi
- Department of Virology, School of Public HealthTehran University of Medical SciencesTehranIran
| | | | - Kaveh Sadeghi
- Department of Virology, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Vahid Salimi
- Department of Virology, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Ahmad Nejati
- Department of Virology, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Talat Mokhtari Azad
- Department of Virology, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Jila Yavarian
- Department of Virology, School of Public HealthTehran University of Medical SciencesTehranIran
- Research Center for Antibiotic Stewardship and Antimicrobial ResistanceTehran University of Medical SciencesTehranIran
| |
Collapse
|
122
|
Hasan A, Rahim R, Nakayama EE, Uno K, Hasan N, Rahman M, Shioda T. Enhancement of IL-6 Production Induced by SARS-CoV-2 Nucleocapsid Protein and Bangladeshi COVID-19 Patients' Sera. Viruses 2023; 15:2018. [PMID: 37896795 PMCID: PMC10611338 DOI: 10.3390/v15102018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a respiratory tract infection caused by severe acute respiratory syndrome coronavirus 2 that can have detrimental effects on multiple organs and accelerate patient mortality. This study, which encompassed 130 confirmed COVID-19 patients who were assessed at three different time points (i.e., 3, 7, and 12 days) after the onset of symptoms, investigated interleukin-6 (IL-6) enhancement induced by a viral nucleocapsid (N) protein from a myeloid cell line. Disease severity was categorized as mild, moderate, or severe. The severe cases were characterized as having significant elevations in serum IL-6, C-reactive protein, D-dimer, ferritin, creatinine, leukocytes, and neutrophil-to-lymphocyte ratio and decreased hemoglobin, hematocrit, and albumin levels compared with mild and moderate cases. To evaluate IL-6-inducing activity, heat-inactivated sera from these patients were incubated with and without the N protein. The findings showed a progressive increase in IL-6 production in severe cases upon N protein stimulation. There was a strong correlation between anti-N antibodies and levels of IL-6 secreted by myeloid cells in the presence of N protein and sera, indicating the crucial role that the anti-N antibody plays in inducing IL-6 production. Uncontrolled IL-6 production played a pivotal role in disease pathogenesis, exacerbating both disease severity and mortality. Efficiently targeting the N protein could potentially be employed as a therapeutic strategy for regulating the immune response and alleviating inflammation in severe cases.
Collapse
Affiliation(s)
- Abu Hasan
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka 1229, Bangladesh; (A.H.); (R.R.); (N.H.)
| | - Rummana Rahim
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka 1229, Bangladesh; (A.H.); (R.R.); (N.H.)
| | - Emi E. Nakayama
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0781, Japan;
| | - Kazuko Uno
- IFN & Host-Defense Research Laboratory, Louis Pasteur Center for Medical Research, Kyoto 606-8225, Japan;
| | - Nazmul Hasan
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka 1229, Bangladesh; (A.H.); (R.R.); (N.H.)
| | - Mizanur Rahman
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka 1229, Bangladesh; (A.H.); (R.R.); (N.H.)
| | - Tatsuo Shioda
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0781, Japan;
| |
Collapse
|
123
|
Piluso M, Ferrari C, Pagani S, Usai P, Raschi S, Parachini L, Oggionni E, Melacini C, D’Arcangelo F, Cattaneo R, Bonacina C, Bernareggi M, Bencini S, Nadalin M, Borelli M, Bellini R, Salandini MC, Scarpazza P. COVID-19 Acute Respiratory Distress Syndrome: Treatment with Helmet CPAP in Respiratory Intermediate Care Unit by Pulmonologists in the Three Italian Pandemic Waves. Adv Respir Med 2023; 91:383-396. [PMID: 37736976 PMCID: PMC10514851 DOI: 10.3390/arm91050030] [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: 08/08/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023]
Abstract
COVID-19 Acute Respiratory Distress Syndrome (CARDS) is the most serious complication of COVID-19. The SARS-CoV-2 outbreaks rapidly saturated intensive care unit (ICU), forcing the application of non-invasive respiratory support (NIRS) in respiratory intermediate care unit (RICU). The primary aim of this study is to compare the patients' clinical characteristics and outcomes (Helmet-Continuous Positive Airway Pressure (H-CPAP) success/failure and survival/death). The secondary aim is to evaluate and detect the main predictors of H-CPAP success and survival/death. A total of 515 patients were enrolled in our observational prospective study based on CARDS developed in RICU during the three Italian pandemic waves. All selected patients were treated with H-CPAP. The worst ratio of arterial partial pressure of oxygen (PaO2) and fraction of inspired oxygen (FiO2) PaO2/FiO2 during H-CPAP stratified the subjects into mild, moderate and severe CARDS. H-CPAP success has increased during the three waves (62%, 69% and 77%, respectively) and the mortality rate has decreased (28%, 21% and 13%). H-CPAP success/failure and survival/death were related to the PaO2/FiO2 (worst score) ratio in H-CPAP and to steroids' administration. D-dimer at admission, FiO2 and positive end expiratory pressure (PEEP) were also associated with H-CPAP success. Our study suggests good outcomes with H-CPAP in CARDS in RICU. A widespread use of steroids could play a role.
Collapse
Affiliation(s)
- Martina Piluso
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Clarissa Ferrari
- Research and Clinical Trials Office, Poliambulanza Foundation Hospital, 25124 Brescia, Italy;
| | - Silvia Pagani
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Pierfranco Usai
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Stefania Raschi
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Luca Parachini
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Elisa Oggionni
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Chiara Melacini
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Francesca D’Arcangelo
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Roberta Cattaneo
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Cristiano Bonacina
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Monica Bernareggi
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Serena Bencini
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Marta Nadalin
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy; (M.N.); (M.B.)
- Cardiothoracic Vascular Department, Respiratory Unit, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Mara Borelli
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy; (M.N.); (M.B.)
- Cardiothoracic Vascular Department, Respiratory Unit, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Roberto Bellini
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Maria Chiara Salandini
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| | - Paolo Scarpazza
- Lung Unit, Cardiothoracic Vascular Department, Vimercate Hospital, 20871 Vimercate, Italy; (M.P.); (P.U.); (S.R.); (L.P.); (E.O.); (C.M.); (F.D.); (R.C.); (C.B.); (M.B.); (S.B.); (R.B.); (M.C.S.); (P.S.)
| |
Collapse
|
124
|
Mohseni Afshar Z, Tavakoli Pirzaman A, Hosseinzadeh R, Babazadeh A, Taghizadeh Moghadam MA, Miri SR, Sio TT, Sullman MJM, Barary M, Ebrahimpour S. Anticoagulant therapy in COVID-19: A narrative review. Clin Transl Sci 2023; 16:1510-1525. [PMID: 37326220 PMCID: PMC10499427 DOI: 10.1111/cts.13569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/17/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can manifest itself in several ways, including coagulopathy and thrombosis. These complications can be the first and sometimes only manifestations of SARS-CoV-2 infection and can occur early or late in the course of the disease. However, these symptoms are more prevalent in hospitalized patients with venous thromboembolism, particularly those admitted to intensive care units. Moreover, various forms of arterial and venous thrombosis, or micro- or macro-vasculature embolisms, have been reported during the current pandemic. They have led to harmful consequences, such as neurological and cardiac events, nearly all resulting from the hypercoagulable state caused by this viral infection. The severe hypercoagulability observed in patients with COVID-19 accounts for most cases of the disease that become critical. Therefore, anticoagulants seem to be one of the most vital therapeutics for treating this potentially life-threatening condition. In the current paper, we present a thorough review of the pathophysiology of COVID-19-induced hypercoagulable state and the use of anticoagulants to treat SARS-CoV-2 infections in different patient groups, as well as their pros and cons.
Collapse
Affiliation(s)
- Zeinab Mohseni Afshar
- Clinical Research Development Center, Imam Reza HospitalKermanshah University of Medical SciencesKermanshahIran
| | | | | | - Arefeh Babazadeh
- Infectious Diseases and Tropical Medicine Research CenterHealth Research Institute, Babol University of Medical SciencesBabolIran
| | | | - Seyed Rouhollah Miri
- Cancer Research CenterCancer Institute of Iran, Tehran University of Medical ScienceTehranIran
| | - Terence T. Sio
- Department of Radiation OncologyMayo ClinicPhoenixArizonaUSA
| | - Mark J. M. Sullman
- Department of Social SciencesUniversity of NicosiaNicosiaCyprus
- Department of Life and Health SciencesUniversity of NicosiaNicosiaCyprus
| | - Mohammad Barary
- Student Research Committee, Virtual School of Medical Education and ManagementShahid Beheshti University of Medical SciencesTehranIran
| | - Soheil Ebrahimpour
- Infectious Diseases and Tropical Medicine Research CenterHealth Research Institute, Babol University of Medical SciencesBabolIran
| |
Collapse
|
125
|
van Blydenstein SA, Omar S, Jacobson B, Menezes CN, Meel R. Right heart echocardiography findings in hypoxic pneumonia patients during the COVID-19 pandemic in a South African population. EUROPEAN HEART JOURNAL. IMAGING METHODS AND PRACTICE 2023; 1:qyad030. [PMID: 39045060 PMCID: PMC11195700 DOI: 10.1093/ehjimp/qyad030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 10/03/2023] [Indexed: 07/25/2024]
Abstract
Aims The right ventricle is affected by Coronavirus disease 19 (COVID-19) via multiple mechanisms, which can result in right ventricular dysfunction (RVD). This study aimed to provide an assessment of right heart function using conventional echocardiography and advanced strain imaging, in patients with hypoxic pneumonia during the COVID-19 pandemic. Methods and results This study was an observational, prospective, single-centre study, including adults with hypoxic pneumonia, in two groups: COVID-19 pneumonia; and non-COVID-19 pneumonia. Bedside echocardiography was performed according to a pre-specified protocol and all right heart measurements were done as per standard guidelines. Right ventricular free wall strain (RVFWS) was measured using Philips® QLAB 11.0 speckle tracking software. Descriptive and comparative statistics were used to analyse data. Spearman Rank Order Correlations were used to determine the correlation between right ventricular (RV) parameters and clinical parameters. Univariate and multivariate logistic regression analyses were performed to characterize the predictors of in-hospital mortality. We enrolled 48 patients with COVID-19 pneumonia and 24 with non-COVID-19 pneumonia. COVID-19 patients were significantly older with a higher frequency of hypertension and diabetes and a trend towards a lower severity of illness score. Mean RVFWS yielded the highest estimates for the prevalence of RVD (81%), with no difference between the two pneumonia groups. Median Tricuspid Annular Plane Systolic Excursion (TAPSE) and right ventricular systolic excursion velocity (RVS') were not significantly different between COVID-19 (TAPSE 17.2 and RVS' 12), and non-COVID-19 pneumonia (TAPSE 17.8 and RVS' 12.1) with P values of 0.29 and 0.86, respectively. Non-COVID-19 pneumonia patients with moderate to severe hypoxaemia (PF < 150) were at greater risk of an elevated RV Systolic Pressure >30 mmHg respiratory rate = 3.25 (CI 1.35-7.82) on admission. Troponin levels discriminated between COVID-19 survivors (6 ng/L) and non-survivors (13 ng/L), P = 0.04. The mortality rate for COVID-19 was high (27%) compared to non-COVID-19 pneumonia (12%). Conclusion Patients with COVID-19 pneumonia had a similar admission prevalence of RVD when compared to patients with non-COVID-19 pneumonia. Despite preserved traditional parameters of RV systolic function, RVFWS was diminished in both groups, and we propose that RVFWS serves as an important marker of the subclinical disease of RV.
Collapse
Affiliation(s)
- S A van Blydenstein
- Division of Pulmonology, Faculty of Health Sciences, Chris Hani Baragwanath Academic Hospital, University of the Witwatersrand, Chris Hani Road, Johannesburg, 1864, South Africa
| | - S Omar
- Division of Critical Care, Faculty of Health Sciences, Chris Hani Baragwanath Academic Hospital, University of the Witwatersrand, Chris Hani Road, Johannesburg, 1864, South Africa
| | - B Jacobson
- Division of Haematology, National Health Laboratory Service, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - C N Menezes
- Division of Infectious Diseases, Faculty of Health Sciences, Chris Hani Baragwanath Academic Hospital, University of the Witwatersrand, Chris Hani Road, Johannesburg, 1864, South Africa
| | - R Meel
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| |
Collapse
|
126
|
Mardani M, Mohammadshahi J, Teimourpour R. Outcomes of COVID-19 in immunocompromised patients: a single center experience. Virusdisease 2023; 34:373-382. [PMID: 37780900 PMCID: PMC10533436 DOI: 10.1007/s13337-023-00832-z] [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: 04/04/2023] [Accepted: 07/18/2023] [Indexed: 10/03/2023] Open
Abstract
Malignancy, bone marrow and organ transplantation are associated with deficient and defective immune systems. Immunocompromised patients are at risk for severe and chronic complication of COVID-19 infection. However, the pathogenesis, diagnosis and management of this comorbidity remain to be elucidated. The purpose of the present study was to describe key aspects of COVID-19 infection in immunocompromised patients. In this retrospective, cross-sectional study, lab findings and outcomes of 418 COVID-19 patients with secondary immunodeficiency disorders admitted to Taleghani Hospital in Tehran, from March 2020 to September 2022 were investigated. Of the 418 immunocompromised patients with COVID-19, 236 (56.5%) were male and the median age of all studied patients was 56.6 ± 16.4 with range of 14 to 92 years. Totally, 198 (47.4%) of the patients died during hospitalization. Remdesivir was used for treatment of all patients. Mortality rate among patients admitted to ICU ward (86.8%) was significantly higher than non ICU admission (p < 0.001). The death rate in patients with CKD was substantially higher than other underlying disease (p < 0.001). In terms of laboratory finding, there was a significant relationship between ICU admission and worse outcome with WBC count (HR = 1.94, 95% CI = 1. 46-2.59, p < 0.001), PMN count (HR = 1.93, 95% CI = 1.452.56, p < 0.001), Hb (HR = 1.49, 95% CI = 1.042.13, p = 0.028), AST (HR = 2.55, 95% CI = 1.913.41, p < 0.001), BUN (HR = 2.56, 95% CI = 2.063.69, p < 0.001), Cr (HR = 2.63, 95% CI = 1.89-3.64, p < 0.001), Comorbidities index (HR = 1.71, 95% CI = 1.29-2.27, p < 0.001) and aging (HR = 1.91, 95% CI = 1.4-2.54, p < 0.001). Immunocompromised status increased the risk of mortality or worse outcome in patients diagnosed with COVID-19. Our finding showed outcome predicting markers in whom the waned immune system encounter new emerging disease and improved our understanding of COVID-19 virus behavior in immunocompromised individuals.
Collapse
Affiliation(s)
- Masoud Mardani
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical, Tehran, Iran
| | - Jafar Mohammadshahi
- Ardabil University of medical science, Ardabil, Iran
- Department of Infectious Diseases, School of Medicine, Ardabil University of Medical Science, Ardabil, Iran
| | - Roghayeh Teimourpour
- Ardabil University of medical science, Ardabil, Iran
- Department of Microbiology, School of Medicine, Ardabil University of Medical Science, Ardabil, Iran
| |
Collapse
|
127
|
Hakami A, Altubayqi T, Qadah EA, Zogel B, Alfaifi SM, Refaei E, Sayed A, Alhazmi L, Sayegh M, Alamer A, Areeshi AS, Hakami D. Biochemical Analysis of Ferritin and D-dimer in COVID-19 Survivors and Non-survivors. Cureus 2023; 15:e45389. [PMID: 37854756 PMCID: PMC10579969 DOI: 10.7759/cureus.45389] [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: 09/17/2023] [Indexed: 10/20/2023] Open
Abstract
Background COVID-19 is a major cause of illness and mortality. The management of COVID-19-related illnesses might change if variables connected to their severity and the requirement for ICU admission could be found. The severity of COVID-19 might be efficiently predicted with several laboratory measures, such as ferritin levels and D-dimer analysis. Objectives This study aimed to evaluate the association between serum D-dimer and ferritin levels and their effects on mortality in patients with COVID-19. Methods This retrospective observational study included all patients with positive real-time polymerase chain reaction (RT-PCR) results for COVID-19 who were hospitalized in the Ministry of Health South Al-Qunfudah General Hospital between March and September 30, 2020. Their laboratory parameters, serum D-dimer, and ferritin levels were evaluated. IBM SPSS Statistics for Windows, Version 26.0 (released 2019; IBM Corp., Armonk, New York, United States) was used to analyze the data. Results A total of 318 COVID-19 patients were analyzed; 56.9% (n=181) were male and 43.1% (n=137) were female. Of these, 78.6% (n=250) survived, including 58% of men and 42% of women. The mean D-dimer was 2.1 mcg/mL (SD=3.16) and the mean ferritin was 698.59 ng/mL (SD=603.11). Non-recovered patients were substantially older (66.16 years old) and had higher D-dimer (5.46) mcg/mL and ferritin levels (992.96) ng/mL. Intubation length and gender did not affect survival. Of the non-survivors, 95.6% (n=239) were admitted to the ICU, and 50% (n=34) required mechanical ventilation. Conclusions COVID-19 infection mortality dramatically increased with older age and increased mean ferritin and plasma D-dimer values, which were significantly higher in COVID-19 non-survivors than in survivors. Therefore, assessing and monitoring these laboratory markers in the early stages of the disease may have a significant impact on preventing disease progression and death.
Collapse
Affiliation(s)
- Abdulrahman Hakami
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Tahani Altubayqi
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Entsar A Qadah
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Basem Zogel
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Samar M Alfaifi
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Eman Refaei
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Ahmed Sayed
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Luai Alhazmi
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Maram Sayegh
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Abdullah Alamer
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Areej S Areeshi
- Department of Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Duaa Hakami
- Department of Medicine, Jazan General Hospital, Jazan Health Affairs, Ministry of Health, Jazan, SAU
| |
Collapse
|
128
|
Jarrahi A, Khodadadi H, Moore NS, Lu Y, Awad ME, Salles EL, Vaibhav K, Baban B, Dhandapani KM. Recombinant human DNase-I improves acute respiratory distress syndrome via neutrophil extracellular trap degradation. J Thromb Haemost 2023; 21:2473-2484. [PMID: 37196848 PMCID: PMC10185489 DOI: 10.1016/j.jtha.2023.04.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/21/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Respiratory failure is the primary cause of death in patients with COVID-19, whereas coagulopathy is associated with excessive inflammation and multiorgan failure. Neutrophil extracellular traps (NETs) may exacerbate inflammation and provide a scaffold for thrombus formation. OBJECTIVES The goal of this study was to determine whether degradation of NETs by recombinant human DNase-I (rhDNase), a safe, Food and Drug Administration-approved drug, reduces excessive inflammation, reverses aberrant coagulation, and improves pulmonary perfusion after experimental acute respiratory distress syndrome (ARDS). METHODS Intranasal poly(I:C), a synthetic double-stranded RNA, was administered to adult mice for 3 consecutive days to simulate a viral infection, and these subjects were randomized to treatment arms, which received either an intravenous placebo or rhDNase. The effects of rhDNase on immune activation, platelet aggregation, and coagulation were assessed in mice and donor human blood. RESULTS NETs were observed in bronchoalveolar lavage fluid and within regions of hypoxic lung tissue after experimental ARDS. The administration of rhDNase mitigated peribronchiolar, perivascular, and interstitial inflammation induced by poly(I:C). In parallel, rhDNase degraded NETs, attenuated platelet-NET aggregates, reduced platelet activation, and normalized the clotting time to improve regional perfusion, as observed using gross morphology, histology, and microcomputed tomographic imaging in mice. Similarly, rhDNase reduced NETs and attenuated platelet activation in human blood. CONCLUSION NETs exacerbate inflammation and promote aberrant coagulation by providing a scaffold for aggregated platelets after experimental ARDS. Intravenous administration of rhDNase degrades NETs and attenuates coagulopathy in ARDS, providing a promising translational approach to improve pulmonary structure and function after ARDS.
Collapse
Affiliation(s)
- Abbas Jarrahi
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Hesam Khodadadi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Nicholas S Moore
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Mohamed E Awad
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Evila L Salles
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA; Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.
| |
Collapse
|
129
|
Lewczuk T, Kazimierczyk R, Sobkowicz B, Lisowska A. Pulmonary embolism in COVID-19 pneumonia patients admitted to temporary hospital - The follow-up study. Adv Med Sci 2023; 68:270-275. [PMID: 37633116 DOI: 10.1016/j.advms.2023.08.002] [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: 04/17/2023] [Revised: 07/04/2023] [Accepted: 08/18/2023] [Indexed: 08/28/2023]
Abstract
PURPOSE Coronavirus disease 2019 (COVID-19) is linked with major coagulation disorders, especially higher risk of developing pulmonary embolism (PE). Our study summarizes COVID-19 patients' management with concomitant PE during the first weeks of pandemic and underlines the importance of D-dimer concentration assessment at admission in terms of prognosis. MATERIAL AND METHODS Study group consisted of 107 outpatients (mean age 68.91 ± 12.83 years) admitted to the Temporary COVID-19 Hospital in Bialystok, Poland with confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and suspicion of PE based on elevated D-dimer concentration (>500 μg/l) and/or low saturation rate (<90%). The clinical follow-up lasted 6 months. Death or re-hospitalization were used as composite clinical endpoint (CEP). RESULTS Cumulative incidence of PE was 62.3% (73/107 patients). Most of the patients were in the intermediate PE risk group according to the pulmonary embolism severity index (PESI) score. The mean total computed tomography (CT) lung involvement of COVID-19 findings was 48.42 ± 27.71%. Neither D-dimers nor NT-proBNP concentrations correlated significantly with the percentage of lung abnormalities in CT. Patients with baseline D-dimer concentration higher than 1429 μg/l had worse prognosis in 6-months observation, log-rank test, p = 0.009. CONCLUSIONS Ongoing SARS-CoV-2 infection along with massive involvement of lung tissue and concomitant thrombi in pulmonary arteries are challenging for physicians. It seems that simple D-dimer concentration assessment at admission may be a helpful tool not only to predict PE but also to estimate the long-term prognosis.
Collapse
Affiliation(s)
- Tomasz Lewczuk
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland
| | | | - Bozena Sobkowicz
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland
| | - Anna Lisowska
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
130
|
Wang Y, Han Q, Zhang S, Xing X, Sun X. New perspective on the immunomodulatory activity of ginsenosides: Focus on effective therapies for post-COVID-19. Biomed Pharmacother 2023; 165:115154. [PMID: 37454595 DOI: 10.1016/j.biopha.2023.115154] [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: 05/12/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
More than 700 million confirmed cases of Coronavirus Disease-2019 (COVID-19) have been reported globally, and 10-60% of patients are expected to exhibit "post-COVID-19 symptoms," which will continue to affect human life and health. In the absence of safer, more specific drugs, current multiple immunotherapies have failed to achieve satisfactory efficacy. Ginseng, a traditional Chinese medicine, is often used as an immunomodulator and has been used in COVID-19 treatment as a tonic to increase blood oxygen saturation. Ginsenosides are the main active components of ginseng. In this review, we summarize the multiple ways in which ginsenosides affect post-COVID-19 symptoms, including inhibition of lipopolysaccharide, tumor necrosis factor signaling, modulation of chemokine receptors and inflammasome activation, induction of macrophage polarization, effects on Toll-like receptors, nuclear factor kappa-B, the mitogen-activated protein kinase pathway, lymphocytes, intestinal flora, and epigenetic regulation. Ginsenosides affect virus-mediated tissue damage, local or systemic inflammation, immune modulation, and other links, thus alleviating respiratory and pulmonary symptoms, reducing the cardiac burden, protecting the nervous system, and providing new ideas for the rehabilitation of patients with post-COVID-19 symptoms. Furthermore, we analyzed its role in strengthening body resistance to eliminate pathogenic factors from the perspective of ginseng-epidemic disease and highlighted the challenges in clinical applications. However, the benefit of ginsenosides in modulating organismal imbalance post-COVID-19 needs to be further evaluated to better validate the pharmacological mechanisms associated with their traditional efficacy and to determine their role in individualized therapy.
Collapse
Affiliation(s)
- Yixin Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China
| | - Qin Han
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China
| | - Shuxia Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China
| | - Xiaoyan Xing
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China.
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China.
| |
Collapse
|
131
|
Zhao J, Xie Y, Meng Z, Liu C, Wu Y, Zhao F, Ma X, Christopher TA, Lopez BJ, Wang Y. COVID-19 and cardiovascular complications: updates of emergency medicine. EMERGENCY AND CRITICAL CARE MEDICINE 2023; 3:104-114. [PMID: 38314258 PMCID: PMC10836842 DOI: 10.1097/ec9.0000000000000095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and SARS-CoV-2 variants, has become a global pandemic resulting in significant morbidity and mortality. Severe cases of COVID-19 are characterized by hypoxemia, hyper-inflammation, cytokine storm in lung. Clinical studies have reported an association between COVID-19 and cardiovascular disease (CVD). Patients with CVD tend to develop severe symptoms and mortality if contracted COVID-19 with further elevations of cardiac injury biomarkers. Furthermore, COVID-19 itself can induce and promoted CVD development, including myocarditis, arrhythmia, acute coronary syndrome, cardiogenic shock, and venous thromboembolism. Although the direct etiology of SARS-CoV-2 induced cardiac injury remains unknown and under-investigated, it is suspected that it is related to myocarditis, cytokine-mediated injury, microvascular injury, and stress-related cardiomyopathy. Despite vaccinations having provided the most effective approach to reducing mortality overall, an adapted treatment paradigm and regular monitoring of cardiac injury biomarkers is critical for improving outcomes in vulnerable populations at risk for severe COVID-19. In this review, we focus on the latest progress in clinic and research on the cardiovascular complications of COVID-19 and provide a perspective of treating cardiac complications deriving from COVID-19 in Emergency Medicine.
Collapse
Affiliation(s)
- Jianli Zhao
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, USA
| | - Yaoli Xie
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zhijun Meng
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
| | - Caihong Liu
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
| | - Yalin Wu
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, USA
| | - Fujie Zhao
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, USA
| | - Xinliang Ma
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Bernard J. Lopez
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
| | - Yajing Wang
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, USA
| |
Collapse
|
132
|
Birlutiu V, Neamtu B, Birlutiu RM, Ghibu AM, Dobritoiu ES. Our Experience with SARS-CoV-2 Infection and Acute Kidney Injury: Results from a Single-Center Retrospective Observational Study. Healthcare (Basel) 2023; 11:2402. [PMID: 37685436 PMCID: PMC10487568 DOI: 10.3390/healthcare11172402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Renal failure in COVID-19 patients is reportedly related to multiple factors such as a direct SARS-CoV-2 cytopathic effect, cytokine storm, the association of pulmonary and/or cardiovascular lesions, the presence of thrombotic microangiopathy, endothelial damage, or the use of potentially nephrotoxic medications. METHODS We retrospectively analyzed 466 cases of SARS-CoV-2 infection, comparing 233 patients with acute kidney injury (AKI) with 233 patients without AKI in terms of their demographic characteristics, comorbidities, clinical background, laboratory investigations, time of AKI onset, therapy, and outcomes after using univariate analysis and a CART decision-tree approach. The latter was constructed in a reverse manner, starting from the top with the root and branching out until the splitting ceased, interconnecting all the predictors to predict the overall outcome (AKI vs. non-AKI). RESULTS There was a statistically significant difference between the clinical form distribution in the two groups, with fewer mild (2 vs. 5) and moderate (54 vs. 133) cases in the AKI group than in the non-AKI group and more severe and critical patients in the AKI cohort (116 vs. 92 and 60 vs. 3). There were four deaths (1.71%) in the non-AKI group and 120 deaths in the AKI group (51.5%) (p-value < 0.001). We noted statistically significant differences between the two study groups in relation to different tissue lesions (LDH), particularly at the pulmonary (CT severity score), hepatic (AST, ALT), and muscular levels (Creatine kinase). In addition, an exacerbated procoagulant and inflammatory profile in the study group was observed. The CART algorithm approach yielded decision paths that helped sort the risk of AKI progression into three categories: the low-risk category (0-40%), the medium-risk category (40-80%), and the high-risk category (>80%). It recognized specific inflammatory and renal biomarker profiles with particular cut-off points for procalcitonin, ferritin, LDH, creatinine, initial urea, and creatinine levels as important predictive factors of AKI outcomes (93.3% overall performance). CONCLUSIONS Our study revealed the association between particular risk factors and AKI progression in COVID-19 patients. Diabetes, dyspnea on admission, the need for supplemental oxygen, and admission to the intensive care unit all had a crucial role in producing unfavorable outcomes, with a death rate of more than 50%. Necessary imaging studies (CT scan severity score) and changes in specific biomarker levels (ferritin and C-reactive protein levels) were also noted. These factors should be further investigated in conjunction with the pathophysiological mechanisms of AKI progression in COVID-19 patients.
Collapse
Affiliation(s)
- Victoria Birlutiu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Romania, Str. Lucian Blaga, Nr. 2A, 550169 Sibiu, Romania
- County Clinical Emergency Hospital, Bvd Corneliu Coposu, Nr. 2-4, 550245 Sibiu, Romania
| | - Bogdan Neamtu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Romania, Str. Lucian Blaga, Nr. 2A, 550169 Sibiu, Romania
- Pediatric Research Department, Pediatric Clinical Hospital Sibiu, Str. Pompeiu Onofreiu, Nr. 2-4, 550166 Sibiu, Romania
| | - Rares-Mircea Birlutiu
- Clinical Hospital of Orthopedics, Traumatology, and Osteoarticular TB Bucharest, B-dul Ferdinand 35–37, Sector 2, 021382 Bucharest, Romania
| | - Andreea Magdalena Ghibu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Romania, Str. Lucian Blaga, Nr. 2A, 550169 Sibiu, Romania
- County Clinical Emergency Hospital, Bvd Corneliu Coposu, Nr. 2-4, 550245 Sibiu, Romania
| | - Elena Simona Dobritoiu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Romania, Str. Lucian Blaga, Nr. 2A, 550169 Sibiu, Romania
- County Clinical Emergency Hospital, Bvd Corneliu Coposu, Nr. 2-4, 550245 Sibiu, Romania
| |
Collapse
|
133
|
Gigante B, Levy JH, van Gorp E, Bartoloni A, Bochaton-Piallat ML, Bäck M, Ten Cate H, Christersson C, Ferreiro JL, Geisler T, Lutgens E, Schulman S, Storey RF, Thachil J, Vilahur G, Liaw PC, Rocca B. Management of patients on antithrombotic therapy with severe infections: a joint clinical consensus statement of the ESC Working Group on Thrombosis, the ESC Working Group on Atherosclerosis and Vascular Biology, and the International Society on Thrombosis and Haemostasis. Eur Heart J 2023; 44:3040-3058. [PMID: 37439553 DOI: 10.1093/eurheartj/ehad388] [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: 10/06/2022] [Revised: 05/01/2023] [Accepted: 05/29/2023] [Indexed: 07/14/2023] Open
Abstract
Patients with severe infections and a pre-existing indication for antithrombotic therapy, i.e. antiplatelet agents, anticoagulant drugs, or their combinations, require integrated clinical counselling among coagulation, infectious disease, and cardiology specialists, due to sepsis-induced coagulopathy that frequently occurs. Bacterial and viral pathogens constitute an increasing threat to global public health, especially for patients with ongoing antithrombotic treatment who have a high risk of thrombotic recurrences and high susceptibility to severe infections with increased morbidity and mortality. Similarly, sepsis survivors are at increased risk for major vascular events. Coagulopathy, which often complicates severe infections, is associated with a high mortality and obligates clinicians to adjust antithrombotic drug type and dosing to avoid bleeding while preventing thrombotic complications. This clinical consensus statement reviews the best available evidence to provide expert opinion and statements on the management of patients hospitalized for severe bacterial or viral infections with a pre-existing indication for antithrombotic therapy (single or combined), in whom sepsis-induced coagulopathy is often observed. Balancing the risk of thrombosis and bleeding in these patients and preventing infections with vaccines, if available, are crucial to prevent events or improve outcomes and prognosis.
Collapse
Affiliation(s)
- Bruna Gigante
- Division of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Solnavägen 30. 17164 and Department of Cardiology, Danderyds Hospital, Entrévägen 2, 182 88, Stockholm, Sweden
| | - Jerrold H Levy
- Departments of Anesthesiology, Critical Care, and Surgery (Cardiothoracic), Duke University School of Medicine, Durham, United States; 2301 Erwin Road, Durham, NC 27710, USA
| | - Eric van Gorp
- Department of Viroscience, Erasmus MC, Rotterdam, PO box 2040 [Room Ee1726], 3000 CA Rotterdam, The Netherlands
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine, Infectious Diseases Unit, University of Florence, Largo Brambilla 3, 50100 Florence, Italy
| | - Marie-Luce Bochaton-Piallat
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1 rue Michel-Servet 1, CH-1211 Genève, Switzerland
| | - Magnus Bäck
- Department of Translational Cardiology, Karolinska Institutet and Karolinska University Hospital, CMM L8:01, 171 76 Stockholm, Sweden
- INSERM U1116, University of Lorraine, Nancy University Hospital, 2 rue Jean Lamour, 54505 Vandoeuvre les Nancy Cedex, France
| | - Hugo Ten Cate
- Department of Internal medicine, Thrombosis Expertise Center, Maastricht University Medical Center and CARIM school for cardiovascular diseases, Universiteitsingel 50, PO Box 616, 6200 MD Maastricht, The Netherlands
- Center for Thrombosis and Haemostasis, Gutenberg University Medical Center, Langenbeckstr. 1, Bldg. 403, 55131 Mainz, Germany
| | - Christina Christersson
- Department of Medical Sciences, Cardiology, Uppsala University, Akademiska Sjukhuset, 75185, Uppsala, Sweden
| | - José Luis Ferreiro
- Department of Cardiology and Bio-Heart Cardiovascular Diseases Research Group; Bellvitge University Hospital - Bellvitge Biomedical Research Institute (IDIBELL); CIBERCV; L'Hospitalet de Llobregat, Hospital Duran i Reynals - Edifici Terapèutic - 2a planta Gran Via de l'Hospitalet, 199, 08908 Hospitalet de Llobregat Barcelona -Spain
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Esther Lutgens
- Cardiovascular Medicine, Experimental CardioVascular Immunology Laboratory, Mayo Clinic, 200 First St SW, 55905, Rochester, MN, USA
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany & German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstrasse 9, 80336, Munich, Germany
| | - Sam Schulman
- Department of Medicine and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada HHS - General Hospital 237, Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Obstetrics and Gynecology, I.M. Sechenov First Moscow State Medical University, Bol'shaya Pirogovskaya Ulitsa, 2, стр. 4, Moscow 119435, Russia
| | - Robert F Storey
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Pegasus House, 463a Glossop Road, Sheffield, S10 2QD, UK
| | - Jecko Thachil
- Department of Haematology, Manchester University Hospitals, Oxford road, Manchester, M13 9WL, UK
| | - Gemma Vilahur
- Institut de Recerca Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, CIBERCV, Avda. Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Patricia C Liaw
- Department of Medicine, Thrombosis & Atherosclerosis Research Institute (TaARI), McMaster University, 237 Barton Street East Hamilton, Ontario L8L 2X2, Canada
| | - Bianca Rocca
- Department of Safety and Bioethics, Section on Pharmacology, Catholic University School of Medicine, Largo F. Vito 1, 00168 Rome, Italy
| |
Collapse
|
134
|
Abudouleh E, Alhamlan F, Al-Qahtani AA, Bohol MF, Al Hazzani A, Khorfan K, Alkaff M, Owaidah T, Al-Qahtani AA. Changes in the Fibrinolytic System of Patients Infected with Severe Acute Respiratory Syndrome Coronavirus 2. J Clin Med 2023; 12:5223. [PMID: 37629265 PMCID: PMC10455675 DOI: 10.3390/jcm12165223] [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: 05/21/2023] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 08/27/2023] Open
Abstract
INTRODUCTION In this study, coagulation and fibrinolysis parameters and their association with disease severity were investigated in coronavirus disease (COVID-19) patients. MATERIALS AND METHODS COVID-19 patients (n = 446) admitted to our institute between 21 February 2021 and 17 March 2022, were recruited. Clinical data and staging were collected from all patients. Blood samples were collected and analyzed for several parameters of fibrinolysis and coagulation, including alpha-2-antiplasmin(α2AP) and plasminogen, thrombin activatable fibrinolysis inhibitor (TAFI), tissue plasminogen activator (tPA), plasminogen activator inhibitor-1 (PAI-1), D-dimer, and fibrinogen levels. RESULTS The TAFI, fibrinogen, and tPA levels were significantly higher in participants who died compared to that of patients who recovered (p < 0.001). However, PAI-1, tPA, and TAFI were significantly higher in patients admitted to the ICU than those of the healthy controls (p < 0.001 for PAI-1 and tPA; p = 0.0331 for TAFI). Our results showed that stage C and D COVID-19 patients had significantly higher levels of PAI-1 (p = 0.003). Furthermore, stage D COVID-19 patients had significantly higher tPA and TAFI values (p = 0.003). CONCLUSIONS Hypofibrinolysis was the most prevalent condition among patients with severe COVID-19. In this study, several coagulation markers were elevated, making them suitable prognostic markers for hypofibrinolysis.
Collapse
Affiliation(s)
- Esra’a Abudouleh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (E.A.); (A.A.H.)
| | - Fatimah Alhamlan
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (F.A.); (M.F.B.)
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Arwa A. Al-Qahtani
- Department of Family Medicine, College of Medicine, Al-Imam Mohammad Ibn Saud Islamic University, Riyadh 13317, Saudi Arabia;
| | - Marie Fe Bohol
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (F.A.); (M.F.B.)
| | - Amal Al Hazzani
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (E.A.); (A.A.H.)
| | - Khadija Khorfan
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia; (K.K.); (M.A.)
| | - Morad Alkaff
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia; (K.K.); (M.A.)
| | - Tarek Owaidah
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia; (K.K.); (M.A.)
- Department of Pathology, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Ahmed A. Al-Qahtani
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (F.A.); (M.F.B.)
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
135
|
Santoro L, Zaccone V, Falsetti L, Ruggieri V, Danese M, Miro C, Di Giorgio A, Nesci A, D’Alessandro A, Moroncini G, Santoliquido A. Role of Endothelium in Cardiovascular Sequelae of Long COVID. Biomedicines 2023; 11:2239. [PMID: 37626735 PMCID: PMC10452509 DOI: 10.3390/biomedicines11082239] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The global action against coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2 infection, shed light on endothelial dysfunction. Although SARS-CoV-2 primarily affects the pulmonary system, multiple studies have documented pan-vascular involvement in COVID-19. The virus is able to penetrate the endothelial barrier, damaging it directly or indirectly and causing endotheliitis and multi-organ injury. Several mechanisms cooperate to development of endothelial dysfunction, including endothelial cell injury and pyroptosis, hyperinflammation and cytokine storm syndrome, oxidative stress and reduced nitric oxide bioavailability, glycocalyx disruption, hypercoagulability, and thrombosis. After acute-phase infection, some patients reported signs and symptoms of a systemic disorder known as long COVID, in which a broad range of cardiovascular (CV) disorders emerged. To date, the exact pathophysiology of long COVID remains unclear: in addition to the persistence of acute-phase infection mechanisms, specific pathways of CV damage have been postulated, such as persistent viral reservoirs in the heart or an autoimmune response to cardiac antigens through molecular mimicry. The aim of this review is to provide an overview of the main molecular patterns of enduring endothelial activation following SARS-CoV-2 infection and to offer the latest summary of CV complications in long COVID.
Collapse
Affiliation(s)
- Luca Santoro
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Vincenzo Zaccone
- Department of Emergency Medicine, Internal and Sub-Intensive Medicine, Azienda Ospedaliero-Universitaria delle Marche, 60126 Ancona, Italy
| | - Lorenzo Falsetti
- Clinica Medica, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.F.); (G.M.)
| | - Vittorio Ruggieri
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Martina Danese
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Chiara Miro
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Angela Di Giorgio
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Antonio Nesci
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Alessia D’Alessandro
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Gianluca Moroncini
- Clinica Medica, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.F.); (G.M.)
| | - Angelo Santoliquido
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
136
|
Lucijanic M, Tjesic-Drinkovic I, Piskac Zivkovic N, Pastrovic F, Rob Z, Bacevac M, Sedinic Lacko M, Dzambas E, Medic B, Vukoja I, Busic I, Grgurevic I, Luksic I, Barsic B. Incidence, Risk Factors and Mortality Associated with Major Bleeding Events in Hospitalized COVID-19 Patients. Life (Basel) 2023; 13:1699. [PMID: 37629556 PMCID: PMC10455881 DOI: 10.3390/life13081699] [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: 07/17/2023] [Revised: 07/30/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Thromboprophylaxis is a mainstay of treatment of hospitalized COVID-19 patients, due to the high occurrence of thrombotic events. This increases the risk of bleeding. However, data on bleeding events and associated risk factors are scarce. Thus, we aimed to investigate the incidence, predictors and clinical outcomes associated with major bleeding in hospitalized COVID-19 patients. We retrospectively evaluated a cohort of 4014 consecutively hospitalized COVID-19 patients treated in a tertiary-level institution in the period 3/2020-3/2021. Bleeding of any kind was documented in 322 (8%) and major bleeding in 129 (3.2%) patients. A total of 129 (40.1%) bleeding events were present at the time of hospital admission, and 193 (59.9%) occurred during hospitalization. In the multivariate logistic regression analysis, intensive-care-unit treatment (adjusted odds ratio (aOR) 6.55; p < 0.001), atrial fibrillation (aOR 2.55; p = 0.029), higher white-blood-cell count (WBC) (aOR 1.03; p = 0.021), lower hemoglobin (aOR 0.97; p = 0.002) and history of bleeding (aOR 17.39; p < 0.001) were recognized as mutually independent predictors of major bleeding. Major bleeding was significantly associated with increased in-hospital mortality compared to non-major-bleeding patients (59.7% vs. 34.8%, p < 0.001), especially if occurring during hospitalization. Median time from major bleeding to death was 5 days. Bleeding events are frequent in hospitalized COVID-19 patients, with a significant proportion of patients presenting at the time of hospital admission, and others almost universally exposed to anticoagulant and corticosteroid therapies. Major bleeding is associated with high mortality, especially if occurring during hospitalization. The recognition of patients at risk and implementation of timely interventions are of high clinical importance.
Collapse
Affiliation(s)
- Marko Lucijanic
- Hematology Department, University Hospital Dubrava, 10000 Zagreb, Croatia;
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ida Tjesic-Drinkovic
- Department of Gastroenterology, Hepatology and Clinical Nutrition, University Hospital Dubrava, 10000 Zagreb, Croatia
| | | | - Frane Pastrovic
- Department of Gastroenterology, Hepatology and Clinical Nutrition, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Zrinka Rob
- Department of Gastroenterology, Hepatology and Clinical Nutrition, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Mersiha Bacevac
- Department of Emergency and Intensive Care Medicine, University Hospital Dubrava, 10000 Zagreb, Croatia
| | | | - Eleonora Dzambas
- Department of Emergency and Intensive Care Medicine, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Barbara Medic
- Department of Gastroenterology, Hepatology and Clinical Nutrition, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Ivan Vukoja
- Gastroenterology and Nephrology Department, General County Hospital Pozega, 34000 Pozega, Croatia
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Iva Busic
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ivica Grgurevic
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Gastroenterology, Hepatology and Clinical Nutrition, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Ivica Luksic
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Maxillofacial Surgery, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Bruno Barsic
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
137
|
Shrestha MR, Basnet A, Tamang B, Khadka S, Maharjan R, Maharjan R, Chand AB, Thapa S, Rai SK. Analysis of altered level of blood-based biomarkers in prognosis of COVID-19 patients. PLoS One 2023; 18:e0287117. [PMID: 37540679 PMCID: PMC10403103 DOI: 10.1371/journal.pone.0287117] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/27/2023] [Indexed: 08/06/2023] Open
Abstract
INTRODUCTION Immune and inflammatory responses developed by the patients with Coronavirus Disease 2019 (COVID-19) during rapid disease progression result in an altered level of biomarkers. Therefore, this study aimed to analyze levels of blood-based biomarkers that are significantly altered in patients with COVID-19. METHODS A cross-sectional study was conducted among COVID-19 diagnosed patients admitted to the tertiary care hospital. Several biomarkers-biochemical, hematological, inflammatory, cardiac, and coagulatory-were analyzed and subsequently tested for statistical significance at P<0.01 by using SPSS version 17.0. RESULTS A total of 1,780 samples were analyzed from 1,232 COVID-19 patients (median age 45 years [IQR 33-57]; 788 [63.96%] male). The COVID-19 patients had significantly (99% Confidence Interval, P<0.01) elevated levels of glucose, urea, alanine transaminase (ALT), aspartate aminotransaminase (AST), alkaline phosphatase (ALP), lactate dehydrogenase (LDH), white blood cell (WBC), C-reactive protein (CRP), procalcitonin (PCT), interleukin-6 (IL-6), ferritin, D-Dimer, and creatinine phosphokinase-MB (CPK-MB) compared to the control group. However, the levels of total protein, albumin, and platelets were significantly (P<0.01) lowered in COVID-19 patients compared to the control group. The elevated levels of glucose, urea, WBC, CRP, D-Dimer, and LDH were significantly (P<0.01) associated with in-hospital mortality in COVID-19 patients. CONCLUSIONS Assessing and monitoring the elevated levels of glucose, urea, ALT, AST, ALP, WBC, CRP, PCT, IL-6, ferritin, LDH, D-Dimer, and CPK-MB and the lowered levels of total protein, albumin, and platelet could provide a basis for evaluation of improved prognosis and effective treatment in patients with COVID-19.
Collapse
Affiliation(s)
- Mahendra Raj Shrestha
- Department of Clinical Laboratory, Nepal Armed Police Force Hospital, Kathmandu, Bagmati, Nepal
| | - Ajaya Basnet
- Department of Medical Microbiology, Shi-Gan International College of Science and Technology, Tribhuvan University, Kathmandu, Bagmati, Nepal
- Department of Microbiology, Nepal Armed Police Force Hospital, Kathmandu, Bagmati, Nepal
| | - Basanta Tamang
- Department of Clinical Laboratory, Nepal Armed Police Force Hospital, Kathmandu, Bagmati, Nepal
| | - Sudip Khadka
- Department of Microbiology and Immunology, Stanford University, Palo Alto, California, United States of America
| | - Rajendra Maharjan
- Department of Clinical Laboratory, Nepal Armed Police Force Hospital, Kathmandu, Bagmati, Nepal
| | - Rupak Maharjan
- Department of Clinical Laboratory, Nepal Armed Police Force Hospital, Kathmandu, Bagmati, Nepal
| | - Arun Bahadur Chand
- Department of Clinical Laboratory, KIST Medical College and Teaching Hospital, Lalitpur, Bagmati, Nepal
| | - Suresh Thapa
- Department of Clinical Laboratory, Nepal Armed Police Force Hospital, Kathmandu, Bagmati, Nepal
| | - Shiba Kumar Rai
- Research Department, Nepal Medical College Teaching Hospital, Kathmandu, Bagmati, Nepal
| |
Collapse
|
138
|
Das S, Sharma T, Bhardwaj A, Srivastava RK. COVID-19 induced ARDS: immunopathology and therapeutics. EXPLORATION OF IMMUNOLOGY 2023:255-275. [DOI: 10.37349/ei.2023.00101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/14/2023] [Indexed: 01/03/2025]
Abstract
The coronavirus disease-2019 (COVID-19) pandemic is a significant threat in the modern era. Clinical studies show that the most common symptom of severe COVID-19 is viral pneumonia-induced acute respiratory distress syndrome (ARDS). The underlying mechanisms by which severe respiratory disease syndrome-coronavirus-2 (SARS-CoV-2) results in ARDS and how certain host factors confer an increased risk of developing severe disease remain unknown. Therefore, identifying the distinctive features of this severe and fatal disease and the therapeutic approaches to COVID-19-induced ARDS remains an immediate need to serve as a basis for best practice models of standardized ARDS treatment. This review article aims to comprehensively discuss the immunopathology of ARDS and provides an overview of the precise role of both the innate and adaptive immune system, with emphasis on the current treatment strategies being tested in the COVID-19-induced ARDS patients. This knowledge will supposedly help in revealing further mechanistic insights into understanding COVID-19-induced ARDS.
Collapse
Affiliation(s)
- Sneha Das
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Tamanna Sharma
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Asha Bhardwaj
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rupesh K. Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| |
Collapse
|
139
|
Gupta B, Ahluwalia P, Gupta N, Gupta A. Role of Nebulized Heparin in Clinical Outcome of COVID-19 Patients with Respiratory Symptoms: A Systematic Review. Indian J Crit Care Med 2023; 27:572-579. [PMID: 37636853 PMCID: PMC10452767 DOI: 10.5005/jp-journals-10071-24511] [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: 05/01/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023] Open
Abstract
Coronavirus disease-2019 (COVID-19) is an extremely contagious illness caused by the SARS-CoV-2 virus and has been declared a pandemic by the World Health Organization (WHO). There are currently no particular treatments, however, nebulized heparin has been offered as a viable therapy. The purpose of this systematic review is to assess the efficacy of nebulized heparin in COVID-19 patients with respiratory symptoms. Methods Relevant studies were identified through a systematic search of the PubMed, Medline, Embase, Cochrane Library and Web of Science, and Scopus databases. The search terms included "nebulized heparin," "COVID-19," and "SARS-CoV-2." Studies that evaluated the use of nebulized heparin in COVID-19 patients with respiratory symptoms were included. The rest of the studies along with those that were not published in English were excluded. The systematic review was registered under PROSPERO-CRD42023413927. Observations Five studies have been included in this systematic review. Case reports, case series, observational studies, and randomized controlled trial (RCT) comprised the studies. The patient sample sizes ranged from 2 to 98. The studies assessed the efficacy of nebulized heparin in COVID-19 patients with variable disease severity. The evaluated outcomes included mortality, hospital stay duration, oxygen requirements, and laboratory parameters. Conclusion Based on the clinical studies included in this systematic review, nebulized heparin may be useful in the management of COVID-19. Oxygen saturation was greater, inflammatory indicators were lower, and hospital stays were shorter in these patients. However, the studies had limitations, including inconsistent sample sizes, varying dosages of nebulized heparin, and no control groups. Nebulized heparin in patients with COVID-19 needs to be studied further to determine its safety and effectiveness. How to cite this article Gupta B, Ahluwalia P, Gupta N, Gupta A. Role of Nebulized Heparin in Clinical Outcome of COVID-19 Patients with Respiratory Symptoms: A Systematic Review. Indian J Crit Care Med 2023;27(8):572-579.
Collapse
Affiliation(s)
- Bhavna Gupta
- Department of Anaesthesia, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Pallavi Ahluwalia
- Department of Anaesthesia, Teerthanker Mahaveer Medical College, Moradabad, Uttar Pradesh, India
| | - Nidhi Gupta
- Department of Anesthesia and Critical Care, Doon Medical College, Dehradun, Uttarakhand, India
| | - Anish Gupta
- Department of CTVS, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| |
Collapse
|
140
|
Patel L, Stenzel A, Van Hove C, Sidebottom A, Kethireddy R, Ha N, Beddow D, Manunga J, Qadri G, Kirven J, Skeik N. Outcomes in patients discharged with extended venous thromboembolism prophylaxis after hospitalization with COVID-19. Vasc Med 2023; 28:331-339. [PMID: 37259526 PMCID: PMC10235916 DOI: 10.1177/1358863x231159945] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
BACKGROUND Venous thromboembolism (VTE) is a known complication of coronavirus disease (COVID-19) in patients requiring hospitalization and intensive care. We examined the association between extended pharmacological VTE prophylaxis and outcomes among patients hospitalized with COVID-19. METHODS This was a retrospective cohort study of patients with an index positive SARS-CoV-2 polymerase chain reaction (PCR) test at the time of, or during hospitalization. Patients who were prescribed extended pharmacological VTE prophylaxis were compared against patients who were not. Multivariable logistic regression was used to produce odds ratio (OR) estimates and Cox proportional hazard models for hazard ratios (HR) with 95% CI to examine the association between pharmacological VTE prophylaxis and outcomes of interest. Primary outcomes were 30- and 90-day VTE events. Secondary outcomes included 30- and 90-day mortality, 30-day superficial venous thrombosis (SVT), acute myocardial infarction (MI), acute ischemic stroke, critical limb ischemia, clinically significant bleeding, and inpatient readmissions. RESULTS A total of 1936 patients were included in the study. Among them, 731 (38%) were discharged on extended pharmacological VTE prophylaxis. No significant difference was found in 30- and 90-day VTE events among groups. Patients discharged on extended VTE prophylaxis showed improved survival at 30 (HR: 0.35; 95% CI: 0.21-0.59) and 90 days (HR: 0.36; 95% CI: 0.23-0.55) and reduced inpatient readmission at 30 days (OR: 0.12; 95% CI: 0.04-0.33) when compared to those without. CONCLUSION Patients discharged on extended VTE prophylaxis after hospitalization due to COVID-19 had similar thrombotic events on follow-up. However, use of extended VTE prophylaxis was associated with improved 30- and 90-day survival and reduced risk of 30-day inpatient readmission.
Collapse
Affiliation(s)
- Love Patel
- Department of Internal Medicine, Abbott
Northwestern Hospital, Minneapolis, MN, USA
| | - Ashley Stenzel
- Care Delivery Research, Allina Health,
Minneapolis, MN, USA
| | | | | | - Rajesh Kethireddy
- Department of Internal Medicine, Abbott
Northwestern Hospital, Minneapolis, MN, USA
| | - Ngoc Ha
- Care Delivery Research, Allina Health,
Minneapolis, MN, USA
| | - David Beddow
- Department of Internal Medicine, Mercy
Hospital, Coon Rapids, MN, USA
| | - Jesse Manunga
- Department of Vascular Surgery, Allina
Health Minneapolis Heart Institute, Minneapolis, MN, USA
| | - Ghaziuddin Qadri
- Department of Internal Medicine, Abbott
Northwestern Hospital, Minneapolis, MN, USA
| | - Justin Kirven
- Department of Internal Medicine, Abbott
Northwestern Hospital, Minneapolis, MN, USA
| | - Nedaa Skeik
- Department of Vascular Medicine, Allina
Health Minneapolis Heart Institute, Minneapolis, MN, USA
| |
Collapse
|
141
|
Gaspar V, Silva B, Ambrioso I, Alves C, Alçada M. Right Ventricle Thrombus in a Massive Pulmonary Embolism COVID-19 Patient. Cureus 2023; 15:e43937. [PMID: 37746420 PMCID: PMC10513353 DOI: 10.7759/cureus.43937] [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: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) infection presents with a wild range of clinical manifestations. Increased inflammatory response and thrombotic risk have been described, being pulmonary embolism a potential cause of death in these patients. Pulmonary embolisms with right ventricle thrombus are rare and have higher mortality rates. This case report concerns a rare clinical presentation of a 75-year-old male with a medical history of right renal transplantation 36 years ago, that presented with a ten-day history of asthenia, followed by fever, shortness of breath, and cough since the day before. He was admitted with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pneumonia and respiratory insufficiency. The next morning the patient worsened, he presented with hypotension, tachycardia, severe refractory hypoxemia, and chest pain. Contrast CT showed a massive pulmonary embolism with a right ventricle thrombus, confirmed by an echocardiogram. Anticoagulation and IV fluids were started, and the patient was transferred to the ICU. He developed obstructive shock, so thrombolysis was performed with a full dose of alteplase. The outcome was good with complete recovery. Posterior investigation excluded other causes for pulmonary embolism. The severity of pulmonary parenchymal disease secondary to COVID-19 correlates with thromboembolic complications, which demand a swift response to avoid death. An abrupt deterioration in oxygenation should raise suspicion for PE in COVID-19 patients, and mostly in the presence of hypotension and tachycardia. In our case report, there was a massive pulmonary embolism with a rare right ventricle thrombus that had a good outcome with medical treatment.
Collapse
Affiliation(s)
- Vasco Gaspar
- Internal Medicine Department, Hospital Distrital de Santarém, Santarém, PRT
| | - Bernardo Silva
- Internal Medicine Department, Hospital Distrital de Santarém, Santarém, PRT
| | - Inês Ambrioso
- Internal Medicine Department, Hospital Distrital de Santarém, Santarém, PRT
| | - Cláudia Alves
- Internal Medicine Department, Hospital Distrital de Santarém, Santarém, PRT
| | - Martim Alçada
- Internal Medicine Department, Hospital Distrital de Santarém, Santarém, PRT
| |
Collapse
|
142
|
Wanjari M, Late S, Sharma R, Munjewar P. Long-term pulmonary and extra-pulmonary consequences of COVID-19: A comprehensive review of current evidence and future perspectives. NARRA J 2023; 3:e156. [PMID: 38454970 PMCID: PMC10919748 DOI: 10.52225/narra.v3i2.156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/26/2023] [Indexed: 03/09/2024]
Abstract
The global impact of the coronavirus disease 2019 (COVID-19) pandemic has been significant, affecting countless individuals worldwide. The existence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to considerable levels of illness and mortality. While considerable attention has been devoted to the immediate handling of COVID-19, there is a growing concern about the long-term effects of this disease. The recent studies have brought to light various complexities associated with COVID-19, encompassing both respiratory and non-respiratory problems such as lung scarring, heart diseases, neurological effects, and psychological consequences. The purpose of this review is to provide a comprehensive understanding of the persistent repercussions of COVID-19. It presents a summary of recent studies that have examined the frequency and severity of these complications, as well as an exploration of the potential mechanisms that contribute to their development. Specifically, it delves into the role of immune dysregulation, prolonged inflammation, and dysfunction of blood vessel linings in the origin of these complications. Moreover, the clinical significance of these long-term consequences is discussed, including their potential impact on healthcare systems and society as a whole. Our review highlights the necessity for continuous monitoring and management of patients diagnosed with COVID-19, along with the importance of conducting follow-up studies over an extended period to determine the most effective strategies for prevention and treatment of these complications.
Collapse
Affiliation(s)
- Mayur Wanjari
- Departement of Research and Development, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, India
| | - Sampada Late
- Department of Nursing, Government Hospital Samudrapur, Wardha, India
| | - Ranjana Sharma
- Department of Medical Surgical Nursing, Srimati Radhikabai Meghe Memorial College of Nursing, Datta Meghe Institute of Higher Education & Research, Wardha, India
| | - Pratiksha Munjewar
- Department of Medical Surgical Nursing, Srimati Radhikabai Meghe Memorial College of Nursing, Datta Meghe Institute of Higher Education & Research, Wardha, India
| |
Collapse
|
143
|
Nadeem AUR, Naqvi SM, Chandy KG, Nagineni VV, Nadeem R, Desai S. Effects of Different Anticoagulation Doses on Moderate-to-Severe COVID-19 Pneumonia With Hypoxemia. Cureus 2023; 15:e43389. [PMID: 37700943 PMCID: PMC10495222 DOI: 10.7759/cureus.43389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2023] [Indexed: 09/14/2023] Open
Abstract
Background COVID-19 is a prothrombotic disease that can cause thromboembolism and microthrombi, which could lead to multiorgan failure and death. Since COVID-19 is a relatively new disease, there are guidelines for anticoagulation dosing for COVID-19 patients without consensus on the dosing. We studied the effects of different doses of anticoagulation in hospitalized patients with COVID-19 pneumonia and hypoxemia on any differences in need for high-flow oxygen, mechanical ventilation, and mortality. We also analyzed the patient population who benefited most from anticoagulation. Methodology We performed a retrospective chart review of all patients who were admitted with the diagnosis of COVID-19 infection with positive polymerase chain reaction, pneumonia (confirmed either by chest X-ray or CT chest), and hypoxemia (oxygen saturation of <94%, while on room air). These patients were studied for outcomes (the need for high-flow oxygen, the requirement for mechanical ventilation, and overall mortality) for different doses of anticoagulation (prophylactic, escalated, and therapeutic). Results The sample consists of 132 subjects, predominantly males (116, 87%), with a mean age of 59 years and a standard deviation of 15. About one-third of the participants had diabetes, and more than 50% had hypertension. Additionally, 27 (20.3%) had a history of heart disease, and 70 (53%) of the subjects were admitted to the intensive care unit (ICU) at some point during the study. Among those admitted to the ICU, about 11 (8%) subjects required mechanical ventilation and 16 (12%) passed away during the study. Those who died had higher use of high-flow oxygen, noninvasive mechanical ventilation, and invasive mechanical ventilation and had a longer stay on mechanical ventilation. There was no significant difference in mortality or need for mechanical ventilation for any strategy of anticoagulation. Conclusions Different doses of anticoagulation did not show any statistically significant relationship between the need for mechanical ventilation and mortality. More patients on high-flow oxygen had received escalated doses of anticoagulation as compared to those who were not on high-flow oxygen. Anticoagulation levels did not have any statistically significant effect on overall survival of patients.
Collapse
Affiliation(s)
- Amin Ur Rehman Nadeem
- Department of Critical Care Medicine, James A Lovell Federal Healthcare Center, Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, USA
| | - Syed M Naqvi
- Department of Internal Medicine, Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, USA
| | - Kurian G Chandy
- Department of Internal Medicine, Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, USA
| | | | - Rashid Nadeem
- Department of Critical Care Medicine, Dubai Hospital, Dubai, ARE
| | - Shreya Desai
- Department of Hematology and Oncology, Georgia Cancer Center at Augusta University, Georgia, USA
| |
Collapse
|
144
|
Corneo E, Garbelotto R, Prestes G, Girardi CS, Santos L, Moreira JCF, Gelain DP, Westphal GA, Kupek E, Walz R, Ritter C, Dal-Pizzol F. Coagulation biomarkers and coronavirus disease 2019 phenotyping: a prospective cohort study. Thromb J 2023; 21:80. [PMID: 37507773 PMCID: PMC10375602 DOI: 10.1186/s12959-023-00524-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Because severe acute respiratory syndrome coronarivus 2 (SARS-CoV-2) leads to severe conditions and thrombus formation, evaluation of the coagulation markers is important in determining the prognosis and phenotyping of patients with COVID-19. METHODS In a prospective study that included 213 COVID-19 patients admitted to the intensive care unit (ICU) the levels of antithrombin, C-reactive protein (CRP); factors XI, XII, XIII; prothrombin and D-dimer were measured. Spearman's correlation coefficient was used to assess the pairwise correlations between the biomarkers. Hierarchical and non-hierarchical cluster analysis was performed using the levels of biomarkers to identify patients´ phenotypes. Multivariate binary regression was used to determine the association of the patient´s outcome with clinical variables and biomarker levels. RESULTS The levels of factors XI and XIII were significantly higher in patients with less severe COVID-19, while factor XIII and antithrombin levels were significantly associated with mortality. These coagulation biomarkers were associated with the in-hospital survival of COVID-19 patients over and above the core clinical factors on admission. Hierarchical cluster analysis showed a cluster between factor XIII and antithrombin, and this hierarchical cluster was extended to CRP in the next step. Furthermore, a non-hierarchical K-means cluster analysis was performed, and two phenotypes were identified based on the CRP and antithrombin levels independently of clinical variables and were associated with mortality. CONCLUSION Coagulation biomarkers were associated with in-hospital survival of COVID-19 patients. Lower levels of factors XI, XII and XIII and prothrombin were associated with disease severity, while higher levels of both CRP and antithrombin clustered with worse prognosis. These results suggest the role of coagulation abnormalities in the development of COVID-19 and open the perspective of identifying subgroups of patients who would benefit more from interventions focused on regulating coagulation.
Collapse
Affiliation(s)
- Emily Corneo
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Santa Catarina, Av. Universitária, Criciúma, 1105, Brazil
| | - Rafael Garbelotto
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Santa Catarina, Av. Universitária, Criciúma, 1105, Brazil
- Intensive Care Unit, Hospital São José, Criciúma, SC, Brazil
| | - Gabriele Prestes
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Santa Catarina, Av. Universitária, Criciúma, 1105, Brazil
| | - Carolina Saibro Girardi
- Departamento de Bioquímica, Centro de Estudos Em Estresse Oxidativo, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Lucas Santos
- Departamento de Bioquímica, Centro de Estudos Em Estresse Oxidativo, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Jose Claudio Fonseca Moreira
- Departamento de Bioquímica, Centro de Estudos Em Estresse Oxidativo, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Daniel Pens Gelain
- Departamento de Bioquímica, Centro de Estudos Em Estresse Oxidativo, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | | | - Emil Kupek
- Public Health Department, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Roger Walz
- Center for Applied Neuroscience (CeNAp), Department of Clinical Medicine, University Hospital - Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Cristiane Ritter
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Santa Catarina, Av. Universitária, Criciúma, 1105, Brazil
- Intensive Care Unit, Hospital São José, Criciúma, SC, Brazil
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Santa Catarina, Av. Universitária, Criciúma, 1105, Brazil.
- Intensive Care Unit, Hospital São José, Criciúma, SC, Brazil.
- Clinical Research Center, Hospital São José, Criciúma, SC, Brazil.
| |
Collapse
|
145
|
Pagliarin LG, de Oliveira LM, dos Anjos VNF, de Souza CDBT, Peiter GC, Façanha Wendel C, Dillmann Groto A, Freire de Melo F, Teixeira KN. In silico evidence of Remdesivir action in blood coagulation cascade modulation in COVID-19 treatment. World J Biol Chem 2023; 14:72-83. [PMID: 37547340 PMCID: PMC10401403 DOI: 10.4331/wjbc.v14.i4.72] [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: 12/30/2022] [Revised: 04/30/2023] [Accepted: 07/03/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has demonstrated several clinical manifestations which include not only respiratory system issues but also liver, kidney, and other organ injuries. One of these abnormalities is coagulopathies, including thrombosis and disseminated intravascular coagulation. Because of this, the administration of low molecular weight heparin is required for patients that need to be hospitalized. In addition, Remdesivir is an antiviral that was used against Middle East Acute Respiratory Syndrome, Ebola, Acute Respiratory Syndrome, and other diseases, showing satisfactory results on recovery. Besides, there is evidence suggesting that this medication can provide a better prognosis for patients with COVID-19. AIM To investigate in silico the interaction between Remdesivir and clotting factors, pursuing a possibility of using it as medicine. METHODS In this in silico study, the 3D structures of angiotensin-converting enzyme 2 (ACE2), Factor I (fibrinogen), Factor II (prothrombin), Factor III (thromboplastin), Factor V (proaccelerin), Factor VII (proconvertin), Factor VIII (antihemophilic factor A), Factor IX (antihemophilic factor B), Factor X (Stuart-Prower factor), and Factor XI (precursor of thromboplastin (these structures are technically called receptors) were selected from the Protein Data Bank. The structures of the antivirals Remdesivir and Osetalmivir (these structures are called ligands) were selected from the PubChem database, while the structure of Atazanavir was selected from the ZINC database. The software AutoDock Tools (ADT) was used to prepare the receptors for molecular docking. Ions, peptides, water molecules, and other ones were removed from each ligand, and then, hydrogen atoms were added to the structures. The grid box was delimited and calculated using the same software ADT. A physiological environment with pH 7.4 is needed to make the ligands interact with the receptors, and still the software Marvin sketch® (ChemAxon®) was used to forecast the protonation state. To perform molecular docking, ADT and Vina software was connected. Using PyMol® software and Discovery studio® software from BIOVIA, it was possible to analyze the amino acid residues from receptors that were involved in the interactions with the ligands. Ligand tortions, atoms that participated in the interactions, and the type, strength, and duration of the interactions were also analyzed using those software. RESULTS Molecular docking analysis showed that Remdesivir and ACE2 had an affinity energy of -8.8 kcal/moL, forming a complex with eight hydrogen bonds involving seven atoms of Remdesivir and five amino acid residues of ACE2. Remdesivir and prothrombin had an interaction with six hydrogen bonds involving atoms of the drug and five amino acid residues of the clotting factor. Similar to that, Remdesivir and thromboplastin presented interactions via seven hydrogen bonds involving five atoms of the drug and four residues of the clotting factor. While Remdesivir and Factor V established a complex with seven hydrogen bonds between six antiviral atoms and six amino acid residues from the factor, and Factor VII connected with the drug by four hydrogen bonds, which involved three atoms of the drug and three residues of amino acids of the factor. The complex between Remdesivir and Factor IX formed an interaction via 11 hydrophilic bonds with seven atoms of the drug and seven residues of the clotting factor, plus one electrostatic bond and three hydrophobic interactions. Factor X and Remdesivir had an affinity energy of -9.6 kcal/moL, and the complex presented 10 hydrogen bonds and 14 different hydrophobic interactions which involved nine atoms of the drug and 16 amino acid residues of the clotting factor. The interaction between Remdesivir and Factor XI formed five hydrogen bonds involving five amino acid residues of the clotting factor and five of the antiviral atoms. CONCLUSION Because of the in silico significant affinity, Remdesivir possibly could act in the severe acute respiratory syndrome coronavirus 2 infection blockade by interacting with ACE2 and concomitantly act in the modulation of the coagulation cascade preventing the hypercoagulable state.
Collapse
Affiliation(s)
| | | | | | | | - Gabrielle Caroline Peiter
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular - Setor Palotina, Universidade Federal do Paraná, Palotina 85.950-000, Paraná, Brazil
| | | | | | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista 45.029-094, Bahia, Brazil
| | - Kádima Nayara Teixeira
- Campus Toledo, Universidade Federal do Paraná, Toledo 85.919-899, Paraná, Brazil
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular - Setor Palotina, Universidade Federal do Paraná, Palotina 85.950-000, Paraná, Brazil
| |
Collapse
|
146
|
Yaluri N, Stančáková Yaluri A, Žeňuch P, Žeňuchová Z, Tóth Š, Kalanin P. Cardiac Biomarkers and Their Role in Identifying Increased Risk of Cardiovascular Complications in COVID-19 Patients. Diagnostics (Basel) 2023; 13:2508. [PMID: 37568870 PMCID: PMC10417576 DOI: 10.3390/diagnostics13152508] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/03/2023] [Accepted: 07/17/2023] [Indexed: 08/13/2023] Open
Abstract
Cardiovascular disease (CVD) is a global health concern, causing significant morbidity and mortality. Both lifestyle and genetics influence the development of CVD. It is often diagnosed late, when the treatment options are limited. Early diagnosis of CVD with help of biomarkers is necessary to prevent adverse outcomes. SARS-CoV-2 infection can cause cardiovascular complications even in patients with no prior history of CVD. This review highlights cardiovascular biomarkers, including novel ones, and their applications as diagnostic and prognostic markers of cardiovascular complications related to SARS-CoV-2 infection. Patients with severe SARS-CoV-2 infection were shown to have elevated levels of cardiac biomarkers, namely N-terminal pro-brain natriuretic peptide (NT-pro-BNP), creatine kinase-myocardial band (CK-MB), and troponins, indicating acute myocardial damage. These biomarkers were also associated with higher mortality rates and therefore should be used throughout COVID-19 patient care to identify high-risk patients promptly to optimize their outcomes. Additionally, microRNAs (miRNAs) are also considered as potential biomarkers and predictors of cardiac and vascular damage in SARS-CoV-2 infection. Identifying molecular pathways contributing to cardiovascular manifestations in COVID-19 is essential for development of early biomarkers, identification of new therapeutic targets, and better prediction and management of cardiovascular outcomes.
Collapse
Affiliation(s)
- Nagendra Yaluri
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | | | - Pavol Žeňuch
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Zuzana Žeňuchová
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Štefan Tóth
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Peter Kalanin
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| |
Collapse
|
147
|
Dong Y, Luo S, Wang Y, Shi Y. Retrospective analysis of COVID-19 among 391 hospitalized patients in the Henan province of China. Medicine (Baltimore) 2023; 102:e34325. [PMID: 37478231 PMCID: PMC10662902 DOI: 10.1097/md.0000000000034325] [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: 04/25/2023] [Accepted: 06/22/2023] [Indexed: 07/23/2023] Open
Abstract
This study investigated the clinical characteristics and risk factors of coronavirus disease 2019 (COVID-19) in patients in designated hospitals (Port Hospital) in the Henan province. A total of 391 COVID-19 patients with complete case information from August 6, 2021 to February 26, 2022 were selected. Logistic regression was used to analyze the differences between the clinical types, ages, and sex of the patients. Multivariate regression analysis of the severe group indicated that underlying diseases [odds ratio (OR):6.76, 95% confidence interval (CI):1.83-24.93], increased urea levels (OR: 1.41, 95% CI: 1.04-1.91), old age (OR: 1.05, 95% CI: 1.00-1.10), and increased lactic dehydrogenase (OR: 1.02, 95% CI: 1.01-1.03) levels and decreased hemoglobin (OR: 0.95, 95% CI: 0.91-1.00) levels were predictors of illness severity. Multivariate regression analysis for those > 50 years of age showed that underlying diseases (OR: 7.06, 95% CI: 2.79-17.89) and increased urea (OR: 1.91, 95% CI: 1.47-2.48), total bilirubin (OR: 1.14, 95% CI: 1.08-1.21), total protein (OR: 1.08, 95% CI: 1.00-1.17), and lactic dehydrogenase (OR: 1.01, 95% CI: 1.00-1.02) levels and decreased albumin (OR: 0.66, 95% CI: 0.58-0.76) levels were characteristics of COVID-19. Multivariate regression analysis stratified by sex showed that the characteristics of COVID-19 patients were increased white blood cell count in males (OR: 0.66, 95% CI: 0.55-0.78) as well as increased creatinine levels (OR: 0.89, 95% CI: 0.87-0.91). This retrospective analysis provides useful information to support the clinical management of patients with COVID-19.
Collapse
Affiliation(s)
- Yang Dong
- Department of Clinical Pathology, Henan Provincial People's Hospital, Department of Clinical Pathology of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Suyan Luo
- Department of Clinical Microbiology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yali Wang
- Department of Clinical Microbiology, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Yujie Shi
- Department of Clinical Pathology, Henan Provincial People's Hospital, Department of Clinical Pathology of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
148
|
Bruzzese A, Vigna E, Terzi D, Greco S, Martino EA, Vangeli V, Mendicino F, Lucia E, Olivito V, Labanca C, Morelli R, Neri A, Morabito F, Zinno F, Mastroianni A, Gentile M. Safe and Effective Administration of Caplacizumab in COVID-19-Associated Thrombotic Thrombocytopenic Purpura. Hematol Rep 2023; 15:448-453. [PMID: 37489376 PMCID: PMC10366750 DOI: 10.3390/hematolrep15030046] [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: 03/27/2023] [Revised: 04/28/2023] [Accepted: 07/12/2023] [Indexed: 07/26/2023] Open
Abstract
Thrombotic thrombocytopenic purpura (TTP) is a potentially life-threatening, rare acute thrombotic microangiopathy (TMA), caused by a severe ADAMTS13 deficiency. As the COVID-19 pandemic rapidly spread around the globe, much data about the pathogenicity of this virus were published. Soon after the detection of the first cases of COVID-19, it was clear that there was a wide range of COVID coagulopathy manifestations, such as deep venous thrombosis, pulmonary thromboembolism, and thrombotic microangiopathies. In the literature, little data have been reported about the association between TTP and COVID-19, and the treatment of COVID-19-associated TTP is still under debate. Here we present the case of a 46-year-old woman who developed a COVID-associated TTP, successfully treated with plasma exchange (PEX), steroids, and caplacizumab.
Collapse
Affiliation(s)
| | - Ernesto Vigna
- Hematology Unit, Azienda Ospedaliera Annunziata, 87100 Cosenza, Italy
| | - Dario Terzi
- Immunohaematology Section, Annunziata Hospital, 87100 Cosenza, Italy
| | - Sonia Greco
- Infectious & Tropical Diseases Unit, Annunziata Hospital, 87100 Cosenza, Italy
| | | | - Valeria Vangeli
- Infectious & Tropical Diseases Unit, Annunziata Hospital, 87100 Cosenza, Italy
| | | | - Eugenio Lucia
- Hematology Unit, Azienda Ospedaliera Annunziata, 87100 Cosenza, Italy
| | - Virginia Olivito
- Hematology Unit, Azienda Ospedaliera Annunziata, 87100 Cosenza, Italy
| | - Caterina Labanca
- Hematology Unit, Azienda Ospedaliera Annunziata, 87100 Cosenza, Italy
| | - Rosellina Morelli
- Medicine Department, "Annunziata" Hospital of Cosenza, 87100 Cosenza, Italy
| | - Antonino Neri
- Scientific Directorate IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy
| | | | - Francesco Zinno
- Immunohaematology Section, Annunziata Hospital, 87100 Cosenza, Italy
| | - Antonio Mastroianni
- Infectious & Tropical Diseases Unit, Annunziata Hospital, 87100 Cosenza, Italy
| | - Massimo Gentile
- Hematology Unit, Azienda Ospedaliera Annunziata, 87100 Cosenza, Italy
- Department of Pharmacy, Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
149
|
Koeck JA, Dohmen SM, Marx G, Eisert A. Comparison of Drug-Related Problems in COVID-19 and Non-COVID-19 Patients Provided by a German Telepharmacy Service for Rural Intensive Care Units. J Clin Med 2023; 12:4739. [PMID: 37510855 PMCID: PMC10380643 DOI: 10.3390/jcm12144739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/05/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Telepharmacy is used to bridge the persisting shortage of specialist ward-based pharmacists, particularly in intensive care units (ICU). During the coronavirus disease 2019 (COVID-19), pharmacotherapy was rapidly developed, which resulted in multiple changes of guidelines. This potentially led to a differing risk for drug-related problems (DRPs) in ICUs. In this study, DRPs were detected in telepharmacy consultations of a German state-wide telemedicine network for adult patients in rural ICUs. The analysis included ICUs of ten general care hospitals with a total of 514 patients and 1056 consultations. The aim of this retrospective, observational cohort study was to compare and analyze the DRPs resulting from ICU patients with or without COVID-19. Furthermore, known risk groups for severe COVID-19 progression (organ insufficiency [kidney, liver], obesity, sex, and/or older age) were investigated with their non-COVID-19 counterparts. As a result, in both groups patients with acute renal insufficiency and without renal replacement therapy showed a significantly higher risk of being affected by one or more DRPs compared to patients with normal renal function. In COVID-19 patients, the initial recommendation of therapeutic anticoagulation (ATC-code B01AB 'Heparin group') resulted in significantly more DRPs compared to non-COVID-19 patients. Therefore, COVID-19 patients with therapeutic anticoagulation and all ICU patients with renal insufficiency should be prioritized for telepharmacy consultations.
Collapse
Affiliation(s)
- Joachim Andreas Koeck
- Pharmacy Department, Erlangen University Hospital, 91054 Erlangen, Germany
- Department of Intensive Care and Intermediate Care, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Sandra Maria Dohmen
- Department of Intensive Care and Intermediate Care, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Gernot Marx
- Department of Intensive Care and Intermediate Care, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Albrecht Eisert
- Hospital Pharmacy, RWTH Aachen University Hospital, 52074 Aachen, Germany
- Institute of Clinical Pharmacology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| |
Collapse
|
150
|
Fernando MC, Hayes T, Besser M, Falter F. Comparison of Blood and Blood Product Transfusion in COVID-19 and Non-COVID-19 Patients Requiring Extracorporeal Membrane Oxygenation for Severe Respiratory Failure. J Clin Med 2023; 12:4667. [PMID: 37510781 PMCID: PMC10381132 DOI: 10.3390/jcm12144667] [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: 06/29/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
COVID-19 has resulted in an exponential increase in patients with severe respiratory failure requiring extracorporeal membrane oxygenation (ECMO). Patients on ECMO regularly require high volumes of blood and blood products but, so far, there has been no comparison of transfusion requirements between COVID-19 and non-COVID-19. Using electronic patient records at two major UK ECMO centres, Royal Papworth Hospital and University Hospital South Manchester, we reviewed the transfusion requirements of patients requiring ECMO between January 2019 to December 2021. A total of 271 patients, including 168 COVID-19 patients were available for analysis. Since COVID-19 patients spent almost twice as long on ECMO (27.1 vs. 14.16 days, p ≤ 0.0001) we indexed transfusion in both groups to days on ECMO to allow comparison. COVID-19 patients required less red blood cells (RBC) per day (0.408 vs. 0.996, p = 0.0005) but more cryoprecipitate transfusions (0.117 vs. 0.106, p = 0.022) compared to non-COVID-19 patients. COVID-19 patients had more than double the mortality of non-COVID-19 patients (47% vs. 20.4%, p = 0.0001) and those who died during the study period had higher platelet transfusion requirements (p = 0.007) than their non-COVID-19 counterparts. Transfusion requirements and coagulopathy differ between COVID-19 and non-COVID-19 patients. The distinctly different transfusion patterns between the two groups remain difficult to interpret, but further investigations may help explain the haematological aspects of severe COVID-19 infection.
Collapse
Affiliation(s)
- Malindra C Fernando
- Department of Anaesthesia and Intensive Care, Royal Papworth Hospital, Cambridge CB2 0AY, UK
| | - Tim Hayes
- Department of Anaesthesia and Intensive Care, Manchester University Hospitals, Manchester M13 9WL, UK
| | - Martin Besser
- Department of Haematology and Blood Transfusion, Cambridge University Hospitals, Cambridge CB2 0QQ, UK
| | - Florian Falter
- Department of Anaesthesia and Intensive Care, Royal Papworth Hospital, Cambridge CB2 0AY, UK
| |
Collapse
|