1
|
Li J, Li SJ. Chondroitin sulfate binds to main protease of SARS-CoV-2 and efficaciously inhibits its activity. Int J Biol Macromol 2025; 306:141547. [PMID: 40020804 DOI: 10.1016/j.ijbiomac.2025.141547] [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: 11/11/2024] [Revised: 01/27/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is constantly mutating and spreading globally, posing a great threat to people's lives and health. The main protease of SARS-CoV-2 (Mpro, also called 3CLpro) is an attractive drug target for SARS-CoV-2, due to its crucial role in processing the viral replication. Here, we showed that chondroitin sulfate (CS) from pig, cattle and shark efficaciously inhibits Mpro activity of SARS-CoV-2 with half maximal inhibitory concentrations (IC50) of 0.148, 0.121 and 0.119 μM, respectively, through a fluorescence resonance energy transfer (FRET) assay. The inhibition pattern of CSs against Mpro activity is competitive inhibition, with inhibition constants (Ki) of CSs derived from pig, bovine and shark are 0.111, 0.096, and 0.107 μM, respectively, indicating significant inhibitory effects of CSs on Mpro activity. Protein fluorescence quenching demonstrated that porcine, bovine and shark CSs strongly bind to Mpro protein with dissociation constants (KD) of 28.31, 28.47 and 20.66 μM at 25 °C at a physiological condition, respectively, mainly through van der Waals and hydrogen bond interactions. Molecular docking and dynamics analysis provides an insight into structural information of the binding of the CSs with Mpro protein. Our findings suggested that CSs from different origins might be a promising food ingredient for the prevention of the SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jinwen Li
- Department of Condensed Matter and Material Physics, School of Physics Science, Nankai University, Tianjin 300071, PR China.
| | - Shu Jie Li
- Department of Biophysics, School of Physics Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, PR China; Qilu Institute of Technology, Shandong 250200, PR China.
| |
Collapse
|
2
|
Agrawal A, Bajaj S, Bhagat U, Chandna S, Arockiam AD, Chan N, Haroun E, Gupta R, Badwan O, Shekhar S, Kathavarayan Ramu S, Nayar D, Jaber W, Griffin BP, Wang TKM. Intracardiac Thrombus in COVID-19 Inpatients: A Nationwide Study of Incidence, Predictors, and Outcomes. Angiology 2025; 76:441-452. [PMID: 38173053 DOI: 10.1177/00033197231225282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
COronaVIrus Disease-2019 (COVID-19) is associated with a hypercoagulable state. Intracardiac thrombosis is a potentially serious complication but has seldom been evaluated in COVID-19 patients. We assessed the incidence, associated factors, and outcomes of COVID-19 patients with intracardiac thrombosis. In 2020, COVID-19 inpatients were identified from the National Inpatient Sample (NIS) database. Data on clinical characteristics, intracardiac thrombosis, and adverse outcomes were collected. Multivariable logistic regression was used to identify factors associated with intracardiac thrombosis, in-hospital mortality, and morbidities. In 2020, 1,683,785 COVID-19 inpatients (mean age 63.8 years, 32.2% females) were studied. Intracardiac thrombosis occurred in 0.10% (1830) of cases. In-hospital outcomes included 13.2% all-cause mortality, 3.5% cardiovascular mortality, 2.6% cardiac arrest, 4.4% acute coronary syndrome (ACS), 16.1% heart failure, 1.3% stroke, and 28.3% acute kidney injury (AKI). Key factors for intracardiac thrombosis were congestive heart failure history and coagulopathy. Intracardiac thrombosis independently linked to higher risks of all-cause mortality (odds ratio [OR]: 3.32 (2.42-4.54)), cardiovascular mortality (OR: 2.95 (1.96-4.44)), cardiac arrest (OR: 2.04 (1.22-3.43)), ACS (OR: 1.62 (1.17-2.22)), stroke (OR: 3.10 (2.11-4.56)), and AKI (OR: 2.13 (1.68-2.69)), but not heart failure. While rare, intracardiac thrombosis in COVID-19 patients independently raised in-hospital mortality and morbidity risks.
Collapse
Affiliation(s)
- Ankit Agrawal
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Suryansh Bajaj
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Umesh Bhagat
- Department of Hospital Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Sanya Chandna
- Department of Hospital Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Aro Daniela Arockiam
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nicholas Chan
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Elio Haroun
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rahul Gupta
- Lehigh Valley Heart Institute, Lehigh Valley Health Network, Allentown, PA, USA
| | - Osamah Badwan
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shashank Shekhar
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shivabalan Kathavarayan Ramu
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Divya Nayar
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Wael Jaber
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brian P Griffin
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tom Kai Ming Wang
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
3
|
Vasković I, Marković M, Udovičić I, Arsenović L, Stojić M, Ignjatović A, Jovanović D, Nešković V. Effectiveness of different anticoagulation regimens in critically ill patients - experience from COVID 19 patients. Blood Coagul Fibrinolysis 2025; 36:82-89. [PMID: 40053316 DOI: 10.1097/mbc.0000000000001354] [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: 12/16/2024] [Accepted: 02/23/2025] [Indexed: 04/05/2025]
Abstract
This study compared the efficacy of therapeutic anticoagulation guided by anti-Xa levels vs. a D-dimer-based protocol in ICU patients with COVID-19. Given the heightened risk of thrombosis despite anticoagulation therapy in some cases, we hypothesised that anti-Xa measurement improves anticoagulation effectiveness and clinical outcomes in this population. We retrospectively analysed data from ICU patients at COVID Hospital Karaburma between April 2020 and December 2021. The primary outcome was the incidence of failed noninvasive ventilation necessitating intubation. Secondary endpoints included mortality rates, thromboembolic and bleeding complications, and anticoagulation effectiveness assessed by antifactor Xa activity. The analysis included 395 patients - 137 in the anti-Xa group and 258 in the D-dimer group. The D-dimer group showed a higher rate of failed noninvasive ventilation requiring intubation (65.7% vs. 50%, P = 0.009). The overall mortality was 48.3%, significantly higher in the D-dimer group (52.7%) compared to the anti-Xa group (40.1%, P = 0.02). Thromboembolic complications were lower in the anti-Xa group (2.9%) than in the D-dimer group (9.7%, P = 0.014), with no significant difference in bleeding. Following the first LMWH administration, 70.8% of patients had anti-Xa levels below the therapeutic and 11.7% below the prophylactic range. Anti-Xa-guided anticoagulation improves survival and reduces thromboembolic complications compared to D-dimer-based treatment without increasing bleeding risk. This study highlights the potential of the anti-Xa assay in managing anticoagulation in critically ill COVID-19 patients. Our findings provide a foundation for future research on using anti-Xa measurements as a guiding tool to optimise anticoagulation therapy in other critically ill populations.
Collapse
Affiliation(s)
- Igor Vasković
- Clinic for Anesthesiology and Intensive Care, Military Medical Academy
| | - Marija Marković
- Clinic for Anesthesiology and Intensive Care, Military Medical Academy
| | - Ivo Udovičić
- Clinic for Anesthesiology and Intensive Care, Military Medical Academy
| | | | - Mihailo Stojić
- Clinic for Anesthesiology and Intensive Care, Military Medical Academy
| | | | - Dragana Jovanović
- Clinic for Anesthesiology and Intensive Care, Military Medical Academy
| | | |
Collapse
|
4
|
Abou Mansour M, El Rassi C, Sleem B, Borghol R, Arabi M. Thromboembolic Events in the Era of COVID-19: A Detailed Narrative Review. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2025; 2025:3804576. [PMID: 40226433 PMCID: PMC11986918 DOI: 10.1155/cjid/3804576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/14/2025] [Indexed: 04/15/2025]
Abstract
COVID-19, caused by the SARS-CoV-2 virus, is not only characterized by respiratory symptoms but is also associated with a wide range of systemic complications, including significant hematologic abnormalities. This is a comprehensive review of the current literature, using PubMed and Google Scholar, on the pathophysiology and incidence of thromboembolic events in COVID-19 patients and thromboprophylaxis. COVID-19 infection induces a prothrombotic state in patients through the dysregulation of the renin-angiotensin-aldosterone system (RAAS), endothelial dysfunction, elevated von Willebrand factor (vWF), and a dysregulated immune response involving the complement system and neutrophil extracellular traps (NETs). As a result, thromboembolic complications have emerged in COVID-19 cases, occurring more frequently in severe cases and hospitalized patients. These thrombotic events affect both venous and arterial circulation, with increased incidences of deep venous thrombosis (DVT), pulmonary embolism (PE), systemic arterial thrombosis, and myocardial infarction (MI). While DVT and PE are more common, the literature highlights the potential lethal consequences of arterial thromboembolism (ATE). This review also briefly examines the ongoing discussions regarding the use of anticoagulants for the prevention of thrombotic events in COVID-19 patients. While theoretically promising, current studies have yielded varied outcomes: Some suggest potential benefits, whereas others report an increased risk of bleeding events among hospitalized patients. Therefore, further large-scale studies are needed to assess the efficacy and safety of anticoagulants for thromboprophylaxis in COVID-19 patients.
Collapse
Affiliation(s)
- Maria Abou Mansour
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Christophe El Rassi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Bshara Sleem
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Raphah Borghol
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, Division of Pediatric Hematology-Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mariam Arabi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, Division of Pediatric Cardiology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
5
|
Jin E, Li B, Wang X, Yan R, Yan C, Gao Y. Prevalence of antiphospholipid antibodies in COVID-19 patients: A meta-analysis. Vascul Pharmacol 2025; 158:107444. [PMID: 39638272 DOI: 10.1016/j.vph.2024.107444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE In some reports, antiphospholipid antibodies (aPL) prevalence is higher in COVID-19 patients. This study intended to compare aPL prevalence between COVID-19 patients and healthy controls, and differences in aPL types using meta-analysis. METHODS This work retrieved published literature about association between COVID-19 and aPL from Embase, Web of Science, PubMed, and The Cochrane Library databases. The observation group was COVID-19 patients, and the control group was healthy individuals. Outcome measures contained any of following aPLs: classic aPL: anti-cardiolipin antibodies (aCL) and anti-β2-glycoprotein-1 antibodies (Anti-β2GP1); other non-criteria aPL: anti-phosphatidylserine/prothrombin antibodies (aPS/PT) and anti-annexin-V antibodies (AnV). Meta-analysis was done on Review Manager 5.4. RESULTS 10 studies involving 2288 patients were deemed eligible for inclusion. The results of the meta-analysis showed that the prevalence of Classic aPL and Any aPL in the COVID-19 group was significantly higher than in the healthy group (Classic aPL, RR = 2.55, 95 % CI = 1.83-3.55, P < 0.00001; Any aPL, RR = 2.34, 95 % CI = 1.46-3.77, P = 0.0005). Anti-β2GP1 IgA antibodies were the most common aPL in COVID-19 patients, with a significantly higher prevalence than in the healthy group (RR = 4.26, 95 % CI = 2.84-6.40, P < 0.00001). The prevalence of the four types of IgM aPL was significantly higher in the COVID-19 group compared to the healthy group, while there was no significant difference in aPL IgG between the two groups. CONCLUSION The prevalence of aPL in COVID-19 patients was significantly higher than in the healthy control group. IgM aPL was more easily detectable in the early stages of COVID-19 infection, while IgG aPL may be of more concern in the later time points of the immune epidemiology following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Er Jin
- Department of Pulmonary and Critical Care Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310002, Zhejiang Province, China
| | - Bei Li
- Department of Geriatric, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, Zhejiang Province, China
| | - Xiaonan Wang
- Department of Geriatric, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, Zhejiang Province, China
| | - Runlan Yan
- Department of Geriatric, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, Zhejiang Province, China
| | - Chenhong Yan
- Department of Pulmonary and Critical Care Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310002, Zhejiang Province, China
| | - Yue Gao
- Department of Geriatric, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, Zhejiang Province, China; Zhejiang Provincial Key laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Major Chronic Disease in the Elderly, Hangzhou 310006, Zhejiang Province, China.
| |
Collapse
|
6
|
Liu T, Kang H. The risk factors for long term cardiovascular symptoms in patients after coronavirus disease 2019 infection. Ann Med 2024; 56:2407065. [PMID: 39317338 PMCID: PMC11423522 DOI: 10.1080/07853890.2024.2407065] [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: 09/28/2023] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024] Open
Abstract
INTRODUCTION Presently, numerous studies have demonstrated that long-term cardiovascular changes after Coronavirus Disease 2019(COVID-19) infection should be considered. The study was aimed to explore the risk factors for post COVID-19 long-term cardiovascular symptoms. METHODS This retrospective observational cross-sectional study involved 204 COVID-19 patients who were admitted to Yantaishan Hospital from January 1, 2023 to January 31, 2023. Demographic and laboratory data were collected and compared between patients who experienced post COVID-19 long-term cardiovascular symptoms and those who did not. Logistic regression analysis was used to identify the risk factors associated with the occurrence of post COVID-19 long-term cardiovascular symptoms. RESULTS Fifty-two participants presented Post COVID-19 cardiovascular symptoms, while the remaining 152 individuals did not show any such symptoms including chest pain, chest tightness, shortness of breath, palpitations, dyspnea, exercise intolerance, and postural tachycardia syndrome. In comparison to the group without post COVID-19 long-term cardiovascular symptoms, the group with post COVID-19 long-term cardiovascular symptoms exhibited a significantly higher prevalence of anxiety and depression (25.0% vs. 4.6%, p = 0.000), as well as significantly elevated C-reactive protein (42.3 mg/L vs. 20.3 mg/L, p = 0.014) and D-dimer (0.3 mg/L vs. 0.22 mg/L, p = 0.024). Anxiety and depression (odds ratio [OR] = 6.403, 95% confidence interval [CI]:2.180-18.809, p = 0.001), C-reactive protein (OR = 1.009, 95%CI:1.003-1.015, p = 0.006), D-dimer (OR = 1.455, 95%CI:1.004-2.109, p = 0.048), and LDL-C (OR = 1.780, 95%CI:1.043-3.040, p = 0.035) were identified as independent risk factors for post COVID-19 long-term cardiovascular symptoms. CONCLUSION Anxiety and depression, C-reactive protein, D-dimer, and LDL-C levels are associated with the development of post COVID-19 long-term cardiovascular symptoms.
Collapse
Affiliation(s)
- Tingting Liu
- Cardiovascular Medicine Department, Yantaishan Hospital, Yantai, China
| | - Haofei Kang
- Cardiovascular Medicine Department, Yantaishan Hospital, Yantai, China
| |
Collapse
|
7
|
Chen L, Jiang YH, Li MY, Huang B, Yuan L, Wan JH, Qin TY, Zeng TT, Chen QG. The Value of Common Laboratory Markers in Predicting the Severity of COVID-19 Patients. Infect Drug Resist 2024; 17:5037-5047. [PMID: 39559342 PMCID: PMC11572049 DOI: 10.2147/idr.s478798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/26/2024] [Indexed: 11/20/2024] Open
Abstract
Purpose The aim of the present study was to identify more effective laboratory markers to assess the severity of corona virus disease 2019 and predict the progression of the disease by collecting more laboratory markers and variables. Patients and Methods In this study, most risk factors, including epidemiological characteristics, blood cell counts, cytokines, and infection markers, were collected from 126 patients with COVID-19 to assess their predictive value. Results The area under curve (AUC) of Albumin (Alb) to fibrinogen (Fib) ratio (AFR) (0.791), Lactate dehydrogenase (LDH) (0.792), myoglobin (MYO) (0.795), C-reactive protein (CRP) (0.801) and lymphocyte count (0.859) were higher than other markers to distinguish severe from non-severe patients in receiver operating characteristic (ROC) analysis. In the univariate logistic regression analysis, thirty-six out of 46 risk factors, including 34 laboratory markers, were significantly associated with increased odds of severe patients. Multivariate logistic regression analysis showed that the CD19+ lymphocyte count, MYO, LDH, and AFR were associated with increased odds of severe disease. Moreover, Lymphocyte count and AFR levels increased, LDH and CRP levels decreased during hospitalization in recovered severe patients, whereas severe lymphocytopenia and continuously increasing LDH levels were observed in deteriorated patients. AFR level increased and CRP level decreased before the disease worsened in the deteriorated patients; however, when the patients deteriorated, AFR decreased and CRP increased significantly. Conclusion CD19+ lymphocyte count, MYO, LDH, and AFR are independent biomarkers for early identification of severe COVID-19. Lymphocyte count, AFR, LDH, and CRP levels were helpful in predicting the clinical progression of the disease.\.
Collapse
Affiliation(s)
- Lian Chen
- Department of Ultrasound Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Yu-Huan Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Mei-Yong Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Bo Huang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Lei Yuan
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Jin-Hua Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Ting-Yu Qin
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Ting-Ting Zeng
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Qing-Gen Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| |
Collapse
|
8
|
Agrawal A, Bhagat U, Arockiam AD, Haroun E, Ming Wang TK. Reply to Letter to the Editor: "Risk Factors Associated With Fatal Thrombosis in COVID-19" regarding: "Improving Risk Analysis for Fatal Thrombosis in COVID-19: A Call for Targeted Anticoagulation". Heart Lung Circ 2024; 33:e63-e65. [PMID: 39521580 DOI: 10.1016/j.hlc.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Affiliation(s)
- Ankit Agrawal
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Umesh Bhagat
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Aro Daniela Arockiam
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Elio Haroun
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tom Kai Ming Wang
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
9
|
Agrawal A, Bajaj S, Bhagat U, Chandna S, Arockiam AD, El Dahdah J, Haroun E, Gupta R, Shekhar S, Raj K, Nayar D, Bajaj D, Chaudhury P, Griffin BP, Wang TKM. Incidence, Predictors, and Outcomes of Venous and Arterial Thrombosis in COVID-19: A Nationwide Inpatient Analysis. Heart Lung Circ 2024; 33:1563-1573. [PMID: 38942623 DOI: 10.1016/j.hlc.2024.04.167] [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: 10/26/2023] [Revised: 03/30/2024] [Accepted: 04/08/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is known to increase the risk of venous thromboembolism (VTE) and arterial thromboembolism (ATE). However, the incidence, predictors, and outcomes of clinical thrombosis for inpatients with COVID-19 are not well known. This study aimed to enhance our understanding of clinical thrombosis in COVID-19, its associated factors, and mortality outcomes. METHOD Hospitalised adult (≥18 years of age) patients with COVID-19 in 2020 were retrospectively identified from the US National Inpatient Sample database. Clinical characteristics, incident VTE, ATE, and in-hospital mortality outcomes were recorded. Multivariable logistic regression was performed to identify clinical factors associated with thrombosis and in-hospital mortality in COVID-19 inpatients. RESULTS A total of 1,583,135 adult patients with COVID-19 in the year 2020 were identified from the National Inpatient Sample database; patients with thrombosis were 41% females with a mean age of 65.4 (65.1-65.6) years. The incidence of thrombosis was 6.1% (97,185), including VTE at 4.8% (76,125), ATE at 3.0% (47,790), and the in-hospital mortality rate was 13.4% (212,785). Patients with thrombosis were more likely to have respiratory symptoms of COVID-19 (76.7% vs 75%, p<0.001) compared with patients without thrombosis. The main factors associated with overall thrombosis, VTE, and ATE were paralysis, ventilation, solid tumours without metastasis, metastatic cancer, and acute liver failure. Although all thrombosis categories were associated with higher in-hospital mortality for COVID-19 inpatients in univariable analyses (p<0.001), they were not in multivariable analyses-thrombosis (odds ratio [OR] 1.24; 95% confidence interval [CI] 0.90-1.70; p=0.19), VTE (OR 0.70; 95% CI 0.52-1.00; p=0.05), and ATE (OR 1.07; 95% CI 0.92-1.25; p=0.36). CONCLUSIONS The association of COVID-19 with thrombosis and VTE increases with increasing severity of the COVID-19 disease. Risk stratification of thrombosis is crucial in COVID-19 patients to determine the necessity of thromboprophylaxis.
Collapse
Affiliation(s)
- Ankit Agrawal
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Suryansh Bajaj
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Umesh Bhagat
- Department of Hospital Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Sanya Chandna
- Department of Hospital Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Aro Daniela Arockiam
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Joseph El Dahdah
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Elio Haroun
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rahul Gupta
- Lehigh Valley Heart Institute, Lehigh Valley Health Network, Allentown, PA, USA
| | - Shashank Shekhar
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kavin Raj
- Division of Cardiology, Department of Medicine, University of California Riverside School of Medicine, Riverside, CA, USA
| | - Divya Nayar
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Divyansh Bajaj
- Department of Pulmonary, Critical Care, and Sleep Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Pulkit Chaudhury
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brian P Griffin
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tom Kai Ming Wang
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
10
|
Iqbal K, Banga A, Arif TB, Rathore SS, Bhurwal A, Naqvi SKB, Mehdi M, Kumar P, Salklan MM, Iqbal A, Ahmed J, Sharma N, Lal A, Kashyap R, Bansal V, Domecq JP. Anticoagulant use before COVID-19 diagnosis prevent COVID-19 associated acute venous thromboembolism or not: A systematic review and meta-analysis. World J Methodol 2024; 14:92983. [PMID: 39310244 PMCID: PMC11230074 DOI: 10.5662/wjm.v14.i3.92983] [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: 02/15/2024] [Revised: 04/24/2024] [Accepted: 05/11/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Coagulopathy and thromboembolic events are associated with poor outcomes in coronavirus disease 2019 (COVID-19) patients. There is conflicting evidence on the effects of chronic anticoagulation on mortality and severity of COVID-19 disease. AIM To summarize the body of evidence on the effects of pre-hospital anticoagulation on outcomes in COVID-19 patients. METHODS A Literature search was performed on LitCovid PubMed, WHO, and Scopus databases from inception (December 2019) till June 2023 for original studies reporting an association between prior use of anticoagulants and patient outcomes in adults with COVID-19. The primary outcome was the risk of thromboembolic events in COVID-19 patients taking anticoagulants. Secondary outcomes included COVID-19 disease severity, in terms of intensive care unit admission or invasive mechanical ventilation/intubation requirement in patients hospitalized with COVID-19 infection, and mortality. The random effects models were used to calculate crude and adjusted odds ratios (aORs) with 95% confidence intervals (95%CIs). RESULTS Forty-six observational studies met our inclusion criteria. The unadjusted analysis found no association between prior anticoagulation and thromboembolic event risk [n = 43851, 9 studies, odds ratio (OR)= 0.67 (0.22, 2.07); P = 0.49; I 2 = 95%]. The association between prior anticoagulation and disease severity was non-significant [n = 186782; 22 studies, OR = 1.08 (0.78, 1.49); P = 0.64; I 2 = 89%]. However, pre-hospital anticoagulation significantly increased all-cause mortality risk [n = 207292; 35 studies, OR = 1.72 (1.37, 2.17); P < 0.00001; I 2 = 93%]. Pooling adjusted estimates revealed a statistically non-significant association between pre-hospital anticoagulation and thromboembolic event risk [aOR = 0.87 (0.42, 1.80); P = 0.71], mortality [aOR = 0.94 (0.84, 1.05); P = 0.31], and disease severity [aOR = 0.96 (0.72, 1.26); P = 0.76]. CONCLUSION Prehospital anticoagulation was not significantly associated with reduced risk of thromboembolic events, improved survival, and lower disease severity in COVID-19 patients.
Collapse
Affiliation(s)
- Kinza Iqbal
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Akshat Banga
- Department of Internal Medicine, Sawai Man Singh Medical College, Jaipur 302004, India
| | - Taha Bin Arif
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Sawai Singh Rathore
- Department of Internal Medicine, Dr. Sampurnanand Medical College, Jodhpur 342003, Rajasthan, India
| | - Abhishek Bhurwal
- Department of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson School of Medicine, New Brunswick, NJ 08901, United States
| | | | - Muhammad Mehdi
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Pankaj Kumar
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Mitali Madhu Salklan
- Department of Internal Medicine, Pandit Bhagwat Dayal Sharma Post Graduate Institute of Medical Sciences, Rohtak 124001, Haryana, India
| | - Ayman Iqbal
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Jawad Ahmed
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Nikhil Sharma
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| | - Amos Lal
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Rahul Kashyap
- Department of Research, Wellspan Health, York, PA 17403, United States
| | - Vikas Bansal
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| | - Juan Pablo Domecq
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| |
Collapse
|
11
|
Dumea E, Lazar M, Chitu-Tisu CE, Barbu EC, Ion DA. COVID-19 associated pulmonary embolism: clinical, biochemical and CT imaging findings. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2024; 62:307-322. [PMID: 38641909 DOI: 10.2478/rjim-2024-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Indexed: 04/21/2024]
Abstract
INTRODUCTION The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection represented a disruptive pathology that emerged in late 2019 with profound implications ranging from individual health to health systems and world economy. Our study aimed to evaluate clinical, biochemical and computerized tomography (CT) parameters values in determining the severity of pulmonary embolism (PE) associated with COVID-19. METHODS We performed an observational cohort study evaluating demographic, clinical, biochemical, coagulation markers, as well as CT imaging parameters. RESULTS In our study on 186 patients with COVID-19, we found that 31 patients (16,66%) had pulmonary embolism. Significant correlations for the patients with PE were detected in C-reactive protein, lactate dehydrogenase, serum ferritin, IL-6, serum myoglobin, NT-proBNP, D-dimers, serum proteins, transaminases as well as white cell blood counts. Patients with pulmonary embolism had a more severe lung involvement, with thrombi distribution mainly involving the lower lobes. CONCLUSION Early identification of PE is an important step for timely and efficient treatment in the intensive care management of COVID-19 patients. Our study showed that high plasmatic values of lactate dehydrogenase, ferritin, IL-6, white blood cells and D-dimers and low proteins serum levels are strongly linked with COVID-19-associated pulmonary embolism.
Collapse
Affiliation(s)
- Eduard Dumea
- 1Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Mihai Lazar
- 1Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
- 2"Prof. Dr. Matei Bals" National Institute for Infectious Diseases, No. 1, Calistrat Grozovici Street, Sector 2, 021105 Bucharest, Romania
| | - Cristina Emilia Chitu-Tisu
- 1Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Ecaterina Constanta Barbu
- 1Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Daniela Adriana Ion
- 1Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| |
Collapse
|
12
|
Azimi Pirsaraei V, Jozpanahi M, Kamali K, Hamzeloo L, Saeid SP. Anticoagulant Use in COVID-19 Patients: A Longitudinal Study From Zanjan, Iran. Cureus 2024; 16:e66798. [PMID: 39268277 PMCID: PMC11392561 DOI: 10.7759/cureus.66798] [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: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Background The mortality and morbidity of thrombotic events in patients with coronavirus disease 2019 (COVID-19) are increasing worldwide. The clinical impact of prophylactic anticoagulation regimens among these patients in Iran remains unclear. This study aimed to evaluate the use of prophylactic anticoagulants and outcomes among COVID-19 patients admitted to a tertiary referral hospital. Methodology Patients diagnosed with COVID-19 and hospitalized between March 20 and June 20, 2020, were included in this longitudinal study after obtaining informed consent. Demographic and clinical data were collected from the hospital information system and medical records. Outcomes during this period were also evaluated. The data were entered into the preparation checklist and analyzed using SPSS version 24 software (IBM Corp., Armonk, NY, USA), employing chi-square, Fisher's exact, and Mann-Whitney U tests. Results Of the 831 enrolled patients, 51.9% were female, and 10.6% needed to be admitted to the intensive care unit (ICU). The mean age of the patients was 57.16 ± 17.32 years, and the mortality rate was estimated to be 9.4%. Mortality rates were significantly higher at older ages, in men, patients with ICU admission, severe pulmonary involvement, malignancy, airway obstruction, ischemic heart disease, and previous cerebrovascular accidents. ICU admission and mortality were statistically significantly higher in those who received concurrent prophylactic anticoagulants and aspirin than in other individuals. Conclusions Our study demonstrated that administering prophylactic aspirin with or without anticoagulant agents in COVID-19 patients did not reduce mortality rates or ICU transfers. However, it is worth noting that anticoagulant prescription was associated with a decrease in ICU admissions, which could potentially alleviate the significantly higher mortality rates observed among ICU patients in this study. Further research is needed to explore the potential benefits of anticoagulants in COVID-19 treatment.
Collapse
Affiliation(s)
| | - Manizhe Jozpanahi
- Department of Infectious Diseases, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, IRN
| | - Koorosh Kamali
- Department of Public Health, School of Public Health, Zanjan University of Medical Sciences, Zanjan, IRN
| | - Leila Hamzeloo
- Department of Infectious Diseases, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, IRN
| | - Seyedeh Pegah Saeid
- Research Committee/General Medicine, Zanjan University of Medical Sciences, Zanjan, IRN
| |
Collapse
|
13
|
Kim HJ, Jeong S, Song J, Park SJ, Park YJ, Oh YH, Jung J, Park SM. Risk of pulmonary embolism and deep vein thrombosis following COVID-19: a nationwide cohort study. MedComm (Beijing) 2024; 5:e655. [PMID: 39006761 PMCID: PMC11246596 DOI: 10.1002/mco2.655] [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: 08/06/2023] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 07/16/2024] Open
Abstract
Recent studies elucidate that coronavirus disease 2019 (COVID-19) patients may face a higher risk of cardiovascular complications. This study aimed to evaluate association of COVID-19 with the risk of pulmonary embolism (PE) or deep vein thrombosis (DVT). This nationwide population-based retrospective cohort study included Korean adult citizens between January 2021 and March 2022 from the Korea Disease Control and Prevention Agency COVID-19 National Health Insurance Service cohort. The Fine and Gray's regression with all-cause death as a competing event was adopted to evaluate PE and DVT risks after COVID-19. This study included a total of 1,601,835 COVID-19 patients and 14,011,285 matched individuals without COVID-19. The risk of PE (adjusted hazard ratio [aHR], 6.25; 95% confidence interval [CI], 3.67-10.66; p < 0.001) and DVT (aHR, 3.05; 95% CI, 1.75-5.29; p < 0.001) was higher in COVID-19 group in individuals without complete COVID-19 vaccination. In addition, individuals with complete COVID-19 vaccination still had a higher risk of COVID-19-related PE (aHR, 1.48; 95% CI, 1.15-1.88; p < 0.001). However, COVID-19 was not a significant risk factor for DVT among those with complete COVID-19 vaccination. COVID-19 was identified as an independent factor that elevated PE and DVT risks, especially for individuals without complete COVID-19 vaccination.
Collapse
Affiliation(s)
- Hye Jun Kim
- Department of Biomedical Sciences Seoul National University College of Medicine Seoul South Korea
| | - Seogsong Jeong
- Department of Biomedical Informatics Korea University College of Medicine Seoul South Korea
| | - Jihun Song
- Department of Biomedical Sciences Seoul National University College of Medicine Seoul South Korea
| | - Sun Jae Park
- Department of Biomedical Sciences Seoul National University College of Medicine Seoul South Korea
| | - Young Jun Park
- Medical Research Center Genomic Medicine Institute Seoul National University Seoul South Korea
| | - Yun Hwan Oh
- Department of Family Medicine Chung-Ang University Gwangmyeong Hospital Chung-Ang University College of Medicine Gwangmyeong South Korea
| | - Jaehun Jung
- Department of Preventive Medicine Gachon University College of Medicine Incheon South Korea
| | - Sang Min Park
- Department of Biomedical Sciences Seoul National University College of Medicine Seoul South Korea
- Department of Family Medicine Seoul National University Hospital Seoul South Korea
| |
Collapse
|
14
|
Siniscalchi C, Ticinesi A, Nouvenne A, Guerra A, Parise A, Finardi L, Cerundolo N, Prati B, Guida L, Meschi T. Non-Invasive Ventilation Support during Hospitalization for SARS-CoV-2 and the Risk of Venous Thromboembolism. J Clin Med 2024; 13:2737. [PMID: 38792278 PMCID: PMC11122199 DOI: 10.3390/jcm13102737] [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: 04/08/2024] [Revised: 04/19/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Background/Objectives: Although SARS-CoV-2 infection is a significant risk factor for venous thromboembolism (VTE), data on the impact of the use of non-invasive ventilation support (NIVS) to mitigate the risk of VTE during hospitalization are scarce. Methods: Data for 1471 SARS-CoV-2 patients, hospitalized in a single hub during the first pandemic wave, were collected from clinical records, including symptom duration and type, information on lung abnormalities on chest computed tomography (CT), laboratory parameters and the use of NIVS. Determining VTE occurrence during hospital stays was the main endpoint. Results: Patients with VTE (1.8%) had an increased prevalence of obesity (26% vs. 11%), diabetes (41% vs. 21%), higher CHA2DS2VASC score (4, IQR 2-5 vs. 3, IQR 1-4, age- and sex-adjusted, p = 0.021) and cough (65% vs. 44%) and experienced significantly higher rates of NIVS (44% vs. 8%). Using a stepwise multivariate logistic regression model, the prevalence of electrocardiogram abnormalities (odds ratio (OR) 2.722, 95% confidence interval (CI) 1.039-7.133, p = 0.042), cough (OR 3.019, 95% CI 1.265-7.202, p = 0.013), CHA2DS2-VASC score > 3 (OR 3.404, 95% CI 1.362-8.513, p = 0.009) and the use of NIVS (OR 15.530, 95% CI 6.244-38.627, p < 0.001) were independently associated with a risk of VTE during hospitalization. NIVS remained an independent risk factor for VTE even after adjustment for the period of admission within the pandemic wave. Conclusions: Our study suggests that NIVS is a risk factor for VTE during hospitalization in SARS-CoV-2 patients. Future studies should assess the optimal prophylactic strategy against VTE in patients with a SARS-CoV-2 infection candidate to non-invasive ventilatory support.
Collapse
Affiliation(s)
- Carmine Siniscalchi
- General and Specialistic Medicine Department, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy; (A.T.); (A.N.); (A.G.); (A.P.); (L.F.); (N.C.); (B.P.); (L.G.); (T.M.)
| | - Andrea Ticinesi
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy; (A.T.); (A.N.); (A.G.); (A.P.); (L.F.); (N.C.); (B.P.); (L.G.); (T.M.)
| | - Antonio Nouvenne
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy; (A.T.); (A.N.); (A.G.); (A.P.); (L.F.); (N.C.); (B.P.); (L.G.); (T.M.)
| | - Angela Guerra
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy; (A.T.); (A.N.); (A.G.); (A.P.); (L.F.); (N.C.); (B.P.); (L.G.); (T.M.)
| | - Alberto Parise
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy; (A.T.); (A.N.); (A.G.); (A.P.); (L.F.); (N.C.); (B.P.); (L.G.); (T.M.)
| | - Lorenzo Finardi
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy; (A.T.); (A.N.); (A.G.); (A.P.); (L.F.); (N.C.); (B.P.); (L.G.); (T.M.)
| | - Nicoletta Cerundolo
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy; (A.T.); (A.N.); (A.G.); (A.P.); (L.F.); (N.C.); (B.P.); (L.G.); (T.M.)
| | - Beatrice Prati
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy; (A.T.); (A.N.); (A.G.); (A.P.); (L.F.); (N.C.); (B.P.); (L.G.); (T.M.)
| | - Loredana Guida
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy; (A.T.); (A.N.); (A.G.); (A.P.); (L.F.); (N.C.); (B.P.); (L.G.); (T.M.)
| | - Tiziana Meschi
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy; (A.T.); (A.N.); (A.G.); (A.P.); (L.F.); (N.C.); (B.P.); (L.G.); (T.M.)
| |
Collapse
|
15
|
Cai W, Zhao Y, Mallappa S. Scoping Review of Clinical Presentations and Outcomes in Patients with Concomitant COVID-19 Infection and Acute Mesenteric Ischaemia. Viruses 2024; 16:506. [PMID: 38675849 PMCID: PMC11054494 DOI: 10.3390/v16040506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
OBJECTIVES COVID-19 infection confers an increased risk of coagulation dysfunction (1) predisposing to thromboembolism in many anatomical sites including the gastrointestinal tract (GIT) (2). This study investigates the clinical presentation and outcome in patients presenting with concurrent COVID-19 infection and gastrointestinal tract ischaemia. Furthermore, differentiation and comparisons are drawn between those with arterial and venous aetiology for mesenteric ischaemia. METHODS A systematic search was undertaken on EMBASE, PubMed, and MEDLINE. Two independent reviewers screened titles, abstracts, and full-text articles according to the inclusion criteria and extracted relevant data. Data analyses were conducted using Excel®. RESULTS Forty-one studies were included in the data analyses, yielding 44 patients. Twenty-six patients had mesenteric arterial occlusion, sixteen patients had mesenteric venous occlusion, and two patients had both arterial and venous mesenteric occlusion. All patients had concurrent COVID-19 infection. The survival rate in patients with arterial aetiology was 38.5% in contrast to 68.8% in patients with venous aetiology. Twelve patients (29.3%) experienced respiratory symptoms in the community before the onset of gastrointestinal symptoms, and five (12.2%) developed gastrointestinal symptoms during their inpatient stay for COVID-19 pneumonitis. CONCLUSIONS Acute mesenteric ischaemia presents a clinical challenge to diagnose due to its non-specific symptoms. Concurrent COVID-19 infection with its predominant respiratory symptoms adds a further challenge in recognising the non-specific symptoms of mesenteric ischaemia. Our study draws attention to the increased thromboembolic risk posed by COVID-19 infection and the need for a high index of suspicion to aid prompt diagnosis and management of acute mesenteric ischaemia, even in the post-pandemic era.
Collapse
Affiliation(s)
- Wenyi Cai
- East Suffolk and North Essex Foundation Trust, Colchester CO4 5JL, UK
- Colchester General Hospital, Turner Road, Colchester CO4 5JL, UK
| | - Yi Zhao
- Imperial College London School of Medicine, London SW7 2DD, UK;
| | - Sreelakshmi Mallappa
- West Hertfordshire Teaching Hospitals NHS Trust, Hertfordshire WD18 0HB, UK;
- The Hillingdon Hospitals NHS Foundation Trust, Uxbridge UB8 3NN, UK
| |
Collapse
|
16
|
Youness M, Mansour S, Sakr F, Olabi S, Atwi S, Martinez IY, El Khatib S, Hallit S, Salameh P, Malaeb D, Hosseini H. Odds and associated factors for thrombosis development among Lebanese COVID-19 patients: a case-control retrospective study. J Pharm Policy Pract 2024; 17:2319743. [PMID: 38505825 PMCID: PMC10950289 DOI: 10.1080/20523211.2024.2319743] [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] [Indexed: 03/21/2024] Open
Abstract
Background Thromboembolism is reported to be up to 27% in COVID-19 patients due to SARS-CoV-2 infection. Dysregulated systemic inflammation and various patient traits play a vital role in thrombosis progression. Purpose To assess odds and associated factors for thrombosis development among Lebanese COVID-19 patients. Methods This was a case-control retrospective study conducted in January-May 2021. Patients infected with COVID-19 and developed thrombosis were classified as cases and patients who were thrombosis-free identified as control. A questionnaire assessed socio-demographics, clinical parameters, and WHO COVID-19 disease severity. Results Among 267 patients, 26 (9.7%) developed thrombosis and the majority of thrombosis 34.6% was myocardial infarction, and the least (3.8%) was for catheter-related thrombosis. Results showed that the risk of thrombosis development is higher in patients with previous thromboembolic event (OR = 9.160) and previous intake of anti-hypertensive medications at home (OR = 3.116). However, females (OR = 0.330; CI: 0.118-0.925), intake of anticoagulants during hospital admission (OR = 0.126; CI: 0.053-0.300) and non-severe COVID-19 were at lower thrombosis risk (OR = 0.273). Patients who developed thromboembolic events had longer hospital stay (OR = 0.077). Conclusion Patients with COVID-19 and thromboembolism were at higher risk of mortality as compared to patients with COVID-19 but without thromboembolism. The use of anticoagulants significantly reduced the risk for thromboembolism.
Collapse
Affiliation(s)
- Mahmoud Youness
- Research Department, Beirut Cardiac Institute, Beirut, Lebanon
| | - Sara Mansour
- School of Pharmacy, Lebanese International University, Beirut, Lebanon
| | - Fouad Sakr
- School of Pharmacy, Lebanese International University, Beirut, Lebanon
| | - Samer Olabi
- Rafic Hariri University Hospital, Beirut, Lebanon
| | - Sarah Atwi
- Rafic Hariri University Hospital, Beirut, Lebanon
| | | | - Sami El Khatib
- Department of Biomedical Sciences, Lebanese International University, Bekaa, Lebanon
- Center for Applied Mathematics and Bioinformatics (CAMB), Gulf University for Science and Technology, Mubarak Al-Abdullah, Kuwait
| | - Souheil Hallit
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik, Jounieh, Lebanon
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
| | - Pascale Salameh
- INSPECT-LB: Institut National de Santé Publique, Epidémiologie Clinique et Toxicologie, Beirut, Lebanon
- Faculty of Pharmacy, Lebanese University, Hadath, Lebanon
- Department of Primary Care and Population Health, University of Nicosia Medical School, Nicosia, Cyprus
- School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Diana Malaeb
- College of Pharmacy, Gulf Medical University, Ajman, United Arab Emirates
| | - Hassan Hosseini
- Neurology Department, Henri Mondor Hospital, AP-HP, Creteil, France
- UPEC-University Paris-Est, Creteil, France
- RAMSAY SANTÉ, HPPE, Champigny sur Marne, France
| |
Collapse
|
17
|
Ferreira-da-Silva R, Maranhão P, Dias CC, Alves JM, Pires L, Morato M, Polónia JJ, Ribeiro-Vaz I. Assessing medication use patterns by clinical outcomes severity among inpatients with COVID-19: A retrospective drug utilization study. Biomed Pharmacother 2024; 172:116242. [PMID: 38340395 DOI: 10.1016/j.biopha.2024.116242] [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: 12/10/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
PURPOSE This study assessed medication patterns for inpatients at a central hospital in Portugal and explored their relationships with clinical outcomes in COVID-19 cases. METHODS A retrospective study analyzed inpatient medication data, coded using the Anatomical Therapeutic Chemical classification system, from electronic patient records. It investigated the association between medications and clinical severity outcomes such as ICU admissions, respiratory/circulatory support needs, and hospital discharge status, including mortality (identified by ICD-10-CM/PCS codes). Multivariate analyses incorporating demographic data and comorbidities were used to adjust for potential confounders and understand the impact of medication patterns on disease progression and outcomes. RESULTS The analysis of 2688 hospitalized COVID-19 patients (55.3% male, average age 62.8 years) revealed a significant correlation between medication types and intensity and disease severity. Cases requiring ICU admission or ECMO support often involved blood and blood-forming organ drugs. Increased use of nervous system and genitourinary hormones was observed in nonsurvivors. Corticosteroids, like dexamethasone, were common in critically ill patients, while tocilizumab was used in ECMO cases. Medications for the alimentary tract, metabolism, and cardiovascular system, although widely prescribed, were linked to more severe cases. Invasive mechanical ventilation correlated with higher usage of systemic anti-infectives and musculoskeletal medications. Trends in co-prescribing blood-forming drugs with those for acid-related disorders, analgesics, and antibacterials were associated with intensive interventions and worse outcomes. CONCLUSIONS The study highlights complex medication regimens in managing severe COVID-19, underscoring specific drug patterns associated with critical health outcomes. Further research is needed to explore these patterns.
Collapse
Affiliation(s)
- Renato Ferreira-da-Silva
- Porto Pharmacovigilance Centre, Faculty of Medicine of the University of Porto, Porto, Portugal; CINTESIS@RISE, Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal.
| | - Priscila Maranhão
- CINTESIS@RISE, Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Cláudia Camila Dias
- CINTESIS@RISE, Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal; Knowledge Management Unit, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - João Miguel Alves
- CINTESIS@RISE, Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Lígia Pires
- Pulmonology Service, Algarve University Hospital Center, Faro, Portugal; Intensive Care Unit, Algarve Private Hospital, Faro, Portugal
| | - Manuela Morato
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy of the University of Porto, Porto, Portugal; LAQV@REQUIMTE, Faculty of Pharmacy of the University of Porto, Porto, Portugal
| | - Jorge Junqueira Polónia
- Porto Pharmacovigilance Centre, Faculty of Medicine of the University of Porto, Porto, Portugal; CINTESIS@RISE, Department of Medicine, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Inês Ribeiro-Vaz
- Porto Pharmacovigilance Centre, Faculty of Medicine of the University of Porto, Porto, Portugal; CINTESIS@RISE, Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| |
Collapse
|
18
|
Maliha ST, Fatemi R, Araf Y. COVID-19 and the brain: understanding the pathogenesis and consequences of neurological damage. Mol Biol Rep 2024; 51:318. [PMID: 38386201 DOI: 10.1007/s11033-024-09279-x] [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: 09/08/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024]
Abstract
SARS-CoV-2 has been known remarkably since December 2019 as a strain of pathogenic coronavirus. Starting from the earlier stages of the COVID-19 pandemic until now, we have witnessed many cases of neurological damage caused by SARS-CoV-2. There are many studies and research conducted on COVID-19-positive-patients that have found brain-related abnormalities with clear neurological symptoms, ranging from simple headaches to life-threatening strokes. For treating neurological damage, knowing the actual pathway or mechanism of causing brain damage via SARS-CoV-2 is very important. For this reason, we have tried to explain the possible pathways of brain damage due to SARS-CoV-2 with mechanisms and illustrations. The SARS-CoV-2 virus enters the human body by binding to specific ACE2 receptors in the targeted cells, which are present in the glial cells and CNS neurons of the human brain. It is found that direct and indirect infections with SARS-CoV-2 in the brain result in endothelial cell death, which alters the BBB tight junctions. These probable alterations can be the reason for the excessive transmission and pathogenicity of SARS-CoV-2 in the human brain. In this precise review, we have tried to demonstrate the neurological symptoms in the case of COVID-19-positive-patients and the possible mechanisms of neurological damage, along with the treatment options for brain-related abnormalities. Knowing the transmission mechanism of SARS-CoV-2 in the human brain can assist us in generating novel treatments associated with neuroinflammation in other brain diseases.
Collapse
Affiliation(s)
- Sumaiya Tasnim Maliha
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, BRAC University, Dhaka, Bangladesh
| | - Rabeya Fatemi
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh
| | - Yusha Araf
- Department of Biotechnology, Bangladesh Agricultural University, Mymensingh, Bangladesh.
| |
Collapse
|
19
|
Naik N, Patel M, Sen R. Developmental Impacts of Epigenetics and Metabolism in COVID-19. J Dev Biol 2024; 12:9. [PMID: 38390960 PMCID: PMC10885083 DOI: 10.3390/jdb12010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Developmental biology is intricately regulated by epigenetics and metabolism but the mechanisms are not completely understood. The situation becomes even more complicated during diseases where all three phenomena are dysregulated. A salient example is COVID-19, where the death toll exceeded 6.96 million in 4 years, while the virus continues to mutate into different variants and infect people. Early evidence during the pandemic showed that the host's immune and inflammatory responses to COVID-19 (like the cytokine storm) impacted the host's metabolism, causing damage to the host's organs and overall physiology. The involvement of angiotensin-converting enzyme 2 (ACE2), the pivotal host receptor for the SARS-CoV-2 virus, was identified and linked to epigenetic abnormalities along with other contributing factors. Recently, studies have revealed stronger connections between epigenetics and metabolism in COVID-19 that impact development and accelerate aging. Patients manifest systemic toxicity, immune dysfunction and multi-organ failure. Single-cell multiomics and other state-of-the-art high-throughput studies are only just beginning to demonstrate the extent of dysregulation and damage. As epigenetics and metabolism directly impact development, there is a crucial need for research implementing cutting-edge technology, next-generation sequencing, bioinformatics analysis, the identification of biomarkers and clinical trials to help with prevention and therapeutic interventions against similar threats in the future.
Collapse
Affiliation(s)
- Noopur Naik
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Mansi Patel
- Institute of Genomics and Integrative Biology, Delhi 110007, India
| | - Rwik Sen
- Active Motif, Inc., Carlsbad, CA 92008, USA
| |
Collapse
|
20
|
Tutak S, Bartosz P, Burda B, Sztwiertnia P, Białecki J. Femoral nerve palsy as a complication due to COVID-19 coagulopathy and iliopsoas muscle hematoma - case report. BMC Musculoskelet Disord 2023; 24:949. [PMID: 38057812 PMCID: PMC10701933 DOI: 10.1186/s12891-023-07062-w] [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: 08/14/2023] [Accepted: 11/25/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND COVID-19 (Coronavirus disease 2019) pandemic is the main medical problem around the world from the end of 2019. We found until now many symptoms of this disease, but one of the most problematic was thrombosis. Wide recommendation on COVID-19 treatment was pharmacological thromboprophylaxis. In some papers we found that clinicians face the problem of bleeding in those patients. Is still unknown that coronavirus could led to the coagulopathy. CASE PRESENTATION We described case report of patient who with COVID-19 disease present femoral nerve palsy caused by the iliopsoas hematoma. There were no deviations in coaguology parameters, patient got standard thromboprophylaxis, besides above probably COVID-19 was risk factor of hematoma formation. Non-operative treatment was applied, thrombophylaxis was discontinued. In the follow up in the radiological exam we saw reduction of the haematoma and patient report decrease of symptoms. CONCLUSIONS We should assess individually patient with COVID-19 according to thrombosis risk factors. Probably we should be more careful in ordering thrombophylaxis medications in COVID-19 patients.
Collapse
Affiliation(s)
- Sławomir Tutak
- Centre of Postgraduate Medical Education, Orthopedic Department in Otwock, Konarskiego 13, Otwock, 05-400, Poland
| | - Paweł Bartosz
- Centre of Postgraduate Medical Education, Orthopedic Department in Otwock, Konarskiego 13, Otwock, 05-400, Poland.
| | - Bartosz Burda
- Centre of Postgraduate Medical Education, Orthopedic Department in Otwock, Konarskiego 13, Otwock, 05-400, Poland
| | - Paweł Sztwiertnia
- Radiological Department in Otwock, Gruca Orthopaedic and Trauma Teaching Hospital, Otwock, Poland
| | - Jerzy Białecki
- Centre of Postgraduate Medical Education, Orthopedic Department in Otwock, Konarskiego 13, Otwock, 05-400, Poland
| |
Collapse
|
21
|
Izquierdo MB, Romo AMN, Zafra AGS, Garrido JDDLG. Predictors of mortality in patients with COVID-19 by flow cytometry. CLINICAL IMMUNOLOGY COMMUNICATIONS 2023; 3:14-20. [PMID: 38014400 PMCID: PMC9918322 DOI: 10.1016/j.clicom.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 11/29/2023]
Abstract
Despite the great impact of severe acute respiratory syndrome caused by coronavirus 2 (SARS-CoV-2), we still lack techniques that allow us to anticipate the natural history of the disease in order to avoid or shorten the clinical period of the disease. The values of nine cytokines were measured in COVID-19+ patients admitted to the Hospital Universitario Reina Sofía (HURS) using flow cytometry. The cytokines measured are IL-1ß, IL-6, MCP-1, IP-10, IL-10, IL-8, IL-12, IFN-γ and TNF-α. Given the absence of previous studies on cytokine values in healthy patients using the flow cytometry technique, and the low availability of resources in the first waves of COVID-19, a control group was lacking, all resources were employed for monitoring sick patients. However, this study has revealed a greater increase in two specific cytokines, which are also found to be higher than the rest in healthy patients: MCP-1 and IP-10, which are mainly responsible for cytokine storm and post-disease thrombosis.
Collapse
Affiliation(s)
| | - Ana María Navas Romo
- Departamento de Inmunología, Hospital Universitario Reina Sofía, Córdoba, España
| | | | | |
Collapse
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
Steinberg HL, Auer TA, Gebauer B, Kloeckner R, Sieren M, Minko P, Jannusch K, Wildgruber M, Schmidt VF, Pinto Dos Santos D, Dratsch T, Hinrichs JB, Torsello G, Stoehr F, Müller L, Herbstreit F, Forsting M, Schaarschmidt BM. Embolization of active arterial bleeding in COVID-19 patients: A multicenter study. Eur J Radiol 2023; 165:110892. [PMID: 37269571 PMCID: PMC10212795 DOI: 10.1016/j.ejrad.2023.110892] [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: 04/07/2023] [Revised: 05/06/2023] [Accepted: 05/23/2023] [Indexed: 06/05/2023]
Abstract
PURPOSE The purpose of this study was to assess the efficacy of transarterial embolization in COVID-19 patients with an arterial bleeding and to investigate differences between various patient groups concerning survival. METHOD We retrospectively reviewed COVID-19 patients undergoing transarterial embolization due to an arterial bleeding in a multicenter study from April 2020 to July 2022 and analyzed the technical success of embolization and survival rate. 30-day survival between various patient groups was analyzed. The Chi- square test and Fisher's exact test were used for testing association between the categorical variables. RESULTS 53 COVID-19 patients (age: 57.3 ± 14.3 years, 37 male) received 66 angiographies due to an arterial bleeding. The initial embolization was technically successful in 98.1% (52/53). In 20.8% (11/53) of patients, additional embolization was necessary due to a new arterial bleeding. A majority of 58.5% (31/53) had a severe course of COVID-19 infection necessitating ECMO-therapy and 86.8% (46/53) of patients received anticoagulation. 30-day survival rate in patients with ECMO-therapy was significantly lower than without ECMO-therapy (45.2% vs. 86.4%, p = 0.004). Patients with anticoagulation did not have a lower 30-day survival rate than without anticoagulation (58.7% vs. 85.7%, p = 0.23). COVID-19 patients with ECMO-therapy developed more frequently a re-bleeding after embolization than non-ECMO-patients (32.3% vs. 4.5%, p = 0.02). CONCLUSIONS Transarterial embolization is a feasible, safe, and effective procedure in COVID-19 patients with arterial bleeding. ECMO-patients have a lower 30-day survival rate than non-ECMO-patients and have an increased risk for re-bleeding. Treatment with anticoagulation could not be identified as a risk factor for higher mortality.
Collapse
Affiliation(s)
- Hannah L Steinberg
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsmedizin Essen, Germany.
| | - Timo A Auer
- Klinik für Radiologie, Charité Universitätsmedizin Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
| | - Bernhard Gebauer
- Klinik für Radiologie, Charité Universitätsmedizin Berlin, Germany
| | - Roman Kloeckner
- Institut für Interventionelle Radiologie, Universitätsklinikum Schleswig-Holstein, Germany
| | - Malte Sieren
- Institut für Interventionelle Radiologie, Universitätsklinikum Schleswig-Holstein, Germany
| | - Peter Minko
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Düsseldorf, Germany
| | - Kai Jannusch
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Düsseldorf, Germany
| | | | | | - Daniel Pinto Dos Santos
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Köln, Germany; Institut für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Frankfurt, Germany
| | - Thomas Dratsch
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Köln, Germany
| | - Jan B Hinrichs
- Institut für Diagnostische und Interventionelle Radiologie, Medizinische Hochschule Hannover, Germany
| | - Giovanni Torsello
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsmedizin Göttingen, Germany
| | - Fabian Stoehr
- Klinik und Poliklinik für Diagnostische und Interventionelle Radiologie, Universitätsmedizin Mainz, Germany
| | - Lukas Müller
- Klinik und Poliklinik für Diagnostische und Interventionelle Radiologie, Universitätsmedizin Mainz, Germany
| | - Frank Herbstreit
- Klinik für Anästhesiologie und Intensivmedizin, Universitätsmedizin Essen, Germany
| | - Michael Forsting
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsmedizin Essen, Germany
| | | |
Collapse
|
26
|
Bae JK, Seo JS, Shin SK, Kim SJ, Kim JH. Does COVID-19 Infection within 1 Week after Total Knee Arthroplasty Affect Patients' Early Clinical Outcomes? A Matched Case-Control Study. J Clin Med 2023; 12:4496. [PMID: 37445528 DOI: 10.3390/jcm12134496] [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: 05/29/2023] [Revised: 06/25/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Recent studies have reported the impact of previous COVID-19 infection on the early clinical outcome after total knee arthroplasty (TKA). However, the timing of infection before the surgery was not constant and a study on patients with COVID-19 infection within 1 week after TKA is lacking. This study aimed to determine the effect of COVID-19 infection within one week after TKA on the postoperative outcomes and to compare the early clinical outcomes to those who were not infected with COVID-19 before and after surgery. No significant differences were observed between the two groups in terms of clinical outcomes or complications. The length of the hospital stay (LOS) was significantly longer for the COVID-19 group than for the non-COVID-19 group (p < 0.05). The erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) levels were higher for the study group on postoperative days 9 and 12 than for those in the control group (p < 0.05). However, D-dimer levels were not significantly different between the two groups. We should cautiously consider that COVID-19 infection within 1 week after TKA may be associated with increased ESR, CRP levels, and length of hospital stay, but they are not associated with the worsening of early clinical outcomes or the occurrence of complications.
Collapse
Affiliation(s)
- Jung-Kwon Bae
- Department of Orthopedic Surgery, Seoul Medical Center, Seoul 02053, Republic of Korea
| | - Jae-Sung Seo
- Department of Orthopedic Surgery, Seoul Medical Center, Seoul 02053, Republic of Korea
| | - Seong-Kee Shin
- Department of Orthopedic Surgery, Seoul Medical Center, Seoul 02053, Republic of Korea
| | - Seo-Jin Kim
- Department of Orthopedic Surgery, Seoul Medical Center, Seoul 02053, Republic of Korea
| | - Jun-Ho Kim
- Department of Orthopedic Surgery, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| |
Collapse
|
27
|
Luu MN, Dang TP, Vo MCH, Quach DT. Prevalence, causes, medical interventions, and mortality outcome of acute gastrointestinal bleeding among COVID-19 inpatients. Curr Med Res Opin 2023; 39:731-737. [PMID: 37026739 DOI: 10.1080/03007995.2023.2201097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/08/2023]
Abstract
OBJECTIVE This study aimed to evaluate the prevalence, causes, medical interventions, and mortality outcome of acute gastrointestinal bleeding (AGIB) among COVID-19 patients hospitalized during the delta pandemic in Vietnam. METHODS The medical records of COVID-19 patients hospitalized in a tertiary hospital in Vietnam from July to October 2021 were retrospectively collected. Data regarding age, sex, comorbidities, COVID-19 severity, onset time of AGIB, therapeutic interventions for AGIB, and mortality outcome were analyzed. RESULTS Of 1567 COVID-19 inpatients, 56 (3.6%) had AGIB. The independent risk factors for AGIB in COVID-19 inpatients included age (OR = 1.03, 95% CI: 1.01-1.04, p = .003), male sex (OR = 1.86, 95% CI: 1.06-3.26, p = .03), chronic liver disease (OR = 6.21, 95% CI: 2.97-13.00, p < .001), and chronic kidney disease (OR = 2.17, 95% CI: 1.01-4.65, p = .047). Among 34 AGIB patients undergoing endoscopy, upper AGIB was determined in 24 (70.6%) patients. Peptic ulcer disease and hemorrhagic erosive gastritis were the most common causes (64.7%, 22/34). The therapeutic interventions for AGIB included blood transfusion (76.8%, 43/56), endoscopic hemostasis (23.5%, 8/34), and surgery (1.8%, 1/56). The mortality rate in the AGIB group was significantly higher than that in the non-AGIB group (46.4% vs. 27.7%, OR = 2.26, 95% CI: 1.32-3.87, p = .002). However, the majority (76.9%) of deaths in COVID-19 inpatients with AGIB were not bleeding-related. CONCLUSIONS Age, male sex, chronic liver disease, and chronic kidney disease are risk factors for AGIB among COVID-19 inpatients. Peptic ulcer disease is the most common cause. COVID-19 inpatients with AGIB have a higher risk of mortality, but a large percentage of deaths are not bleeding-related.
Collapse
Affiliation(s)
- Mai N Luu
- Department of Internal Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Department of Gastroenterology, Nhan Dan Gia Dinh Hospital, Ho Chi Minh City, Vietnam
| | - Thinh P Dang
- Department of Endoscopy, Nhan Dan Gia Dinh Hospital, Ho Chi Minh City, Vietnam
| | - Minh-Cong H Vo
- Department of Gastroenterology, Nhan Dan Gia Dinh Hospital, Ho Chi Minh City, Vietnam
| | - Duc T Quach
- Department of Internal Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Department of Gastroenterology, Nhan Dan Gia Dinh Hospital, Ho Chi Minh City, Vietnam
| |
Collapse
|
28
|
AlLehaibi LH, Alomar M, Almulhim A, Al-Makki S, Alrwaili NR, Al-Bassam S, Alsultan S, Al Saeed J, Alsheef M, Abraham I, Alamer A. Effectiveness and Safety of Enoxaparin Versus Unfractionated Heparin as Thromboprophylaxis in Hospitalized COVID-19 Patients: Real-World Evidence. Ann Pharmacother 2023; 57:361-374. [PMID: 35942505 PMCID: PMC9996167 DOI: 10.1177/10600280221115299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Coronavirus 2019 (COVID-19) patients are at risk of thrombosis. Literature that compares the effectiveness of enoxaparin to unfractionated heparin (UFH) in COVID-19 patients is scarce. OBJECTIVE We aimed to evaluate the effectiveness and safety of enoxaparin compared with UFH when used at their standard/intermediate dosing in COVID-19 patients. METHODS This was a retrospective study conducted at a large COVID-19 center located in Eastern Province, Saudi Arabia. Confirmed COVID-19 cases (≥18 years old) admitted between January and December 2020 were randomly screened for inclusion. Exclusion criteria were patients receiving therapeutic anticoagulation, on chronic anticoagulation, had active bleeding, a platelet count <25 × 109/L, or an incomplete electronic file. The primary endpoint was the occurrence of any thrombotic event (pulmonary embolism, deep venous thrombosis, stroke, or myocardial infarction) or mortality. Secondary endpoints were major or minor bleeding. We applied inverse propensity score weighting (IPTW) with survival analysis to analyze the primary endpoint. Logistic regression was used for the secondary endpoint. RESULTS A total of 980 patients were included (enoxaparin, n = 470 and UFH, n = 510) with a mean age (±SD) of 47.7 (± 12.3) for the enoxaparin arm and 52 (±13.9) for the UFH arm. There was a statistically significant difference in the primary endpoint with an adjusted hazard ratio (aHR) of 0.46 (95%CI: 0.22 to 0.96, P = 0.039) in favor of the enoxaparin arm. There was no statistically significant difference in major or minor bleeding rates between the two arms. CONCLUSION AND RELEVANCE When compared with UFH, enoxaparin was associated with a significant reduction in thrombotic events or mortality among COVID-19 patients. The results need confirmation from randomized controlled trials.
Collapse
Affiliation(s)
- Lina H. AlLehaibi
- Eastern Health Cluster, Dammam Medical Complex, Dammam, Saudi Arabia
| | - Mukhtar Alomar
- Eastern Health Cluster, Dammam Medical Complex, Dammam, Saudi Arabia
| | - Abdulaziz Almulhim
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Sarah Al-Makki
- Eastern Health Cluster, Dammam Medical Complex, Dammam, Saudi Arabia
| | - Nazar R. Alrwaili
- Department of Pharmacy Practice, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Shahad Al-Bassam
- Department of Pharmacy Practice, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Semat Alsultan
- Department of Pharmacy Practice, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Jenan Al Saeed
- Department of Pharmacy Practice, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohammad Alsheef
- Department of Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ivo Abraham
- Center for Health Outcomes & PharmacoEconomic Research, University of Arizona, Tucson, AZ, USA
| | - Ahmad Alamer
- Center for Health Outcomes & PharmacoEconomic Research, University of Arizona, Tucson, AZ, USA
- Department of Clinical Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| |
Collapse
|
29
|
Sandhu HS, Lambert J, Steckler Z, Park L, Stromberg A, Ramirez J, Yang CFJ. Outpatient medications associated with protection from COVID-19 hospitalization. PLoS One 2023; 18:e0282961. [PMID: 37000808 PMCID: PMC10065249 DOI: 10.1371/journal.pone.0282961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 02/28/2023] [Indexed: 04/01/2023] Open
Abstract
The COVID-19 pandemic remains the pre-eminent global health problem, and yet after more than three years there is still no prophylactic agent against the disease aside from vaccines. The objective of this study was to evaluate whether pre-existing, outpatient medications approved by the US Food and Drug Administration (FDA) reduce the risk of hospitalization due to COVID-19. This was a retrospective cohort study of patients from across the United States infected with COVID-19 in the year 2020. The main outcome was adjusted odds of hospitalization for COVID-19 amongst those positive for the infection. Outcomes were adjusted for known risk factors for severe disease. 3,974,272 patients aged 18 or older with a diagnosis of COVID-19 in 2020 met our inclusion criteria and were included in the analysis. Mean age was 50.7 (SD 18). Of this group, 290,348 patients (7.3%) were hospitalized due to COVID-19, similar to the CDC's reported estimate (7.5%). Four drugs showed protective effects against COVID-19 hospitalization: rosuvastatin (aOR 0.91, p = 0.00000024), empagliflozin-metformin (aOR 0.69, p = 0.003), metformin (aOR 0.97, p = 0.017), and enoxaparin (aOR 0.88, p = 0.0048). Several pre-existing medications for outpatient use may reduce severity of disease and protect against COVID-19 hospitalization. Well-designed clinical trials are needed to assess the efficacy of these agents in a therapeutic or prophylactic setting.
Collapse
Affiliation(s)
- Harpal Singh Sandhu
- Department of Bioengineering, University of Louisville Speed School of Engineering, Louisville, KY, United States of America
| | - Joshua Lambert
- University of Cincinnati College of Nursing, Cincinnati, OH, United States of America
| | - Zach Steckler
- Dr. Bing Zhang Department of Statistics, University of Kentucky, Lexington, KY, United States of America
| | - Lee Park
- Dr. Bing Zhang Department of Statistics, University of Kentucky, Lexington, KY, United States of America
| | - Arnold Stromberg
- Norton Infectious Diseases Institute, Norton Hospital, Louisville, KY, United States of America
| | - Julio Ramirez
- Norton Infectious Diseases Institute, Norton Hospital, Louisville, KY, United States of America
| | - Chi-fu Jeffrey Yang
- Department of Surgery, Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
30
|
Zapata-Cachafeiro M, Prieto-Campo Á, Portela-Romero M, Carracedo-Martínez E, Lema-Oreiro M, Piñeiro-Lamas M, Chaudhuri S, Salgado-Barreira Á, Figueiras A. Effect of Previous Anticoagulant Treatment on Risk of COVID-19. Drug Saf 2023; 46:273-281. [PMID: 36562942 PMCID: PMC9782265 DOI: 10.1007/s40264-022-01266-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Little is known about the role played by anticoagulants in COVID-19. OBJECTIVE The aim of this study was to assess the impact of previous anticoagulant treatment on risk of hospitalization due to COVID-19, progression to severe COVID-19 and susceptibility to COVID-19 infection. METHODS We conducted a multiple population-based case-control study in northwest Spain, in 2020, to assess (1) risk of hospitalization: cases were all patients admitted due to COVID-19 with PCR confirmation, and controls were a random matched sample of subjects without a positive PCR; (2) progression: cases were hospitalized COVID-19 subjects, and controls were all non-hospitalized COVID-19 patients; and (3) susceptibility: cases were patients with a positive PCR (hospitalized and non-hospitalized), and the controls were the same as for the hospitalization model. Adjusted odds ratios (ORs) and 95% confidence intervals (95% CIs) were calculated using a generalized linear mixed model. RESULTS The consumption of antivitamin K and direct-acting anticoagulants apparently was not associated with the risk of progression to severe COVID-19 (OR 0.93 [95% CI 0.74-1.17] and OR 1.04 [95% CI 0.79-1.36], respectively). Antivitamin K anticoagulants were associated with a significantly lower risk of hospitalization (OR 0.77 [95% CI 0.64-0.93]), which, in part, can be explained by a decreased risk of susceptibility to infection (OR 0.83 [95% CI 0.74-0.92]). The use of direct-acting anticoagulants was not associated with the risk of hospitalization, although it also seems to decrease susceptibility (OR 0.85 [95% CI 0.74-0.98]). It has also been observed that low-molecular-weight heparins were associated with an increased risk of progression to severe COVID-19 (OR 1.25 [95% CI 1.01-1.55]). CONCLUSION The results of this study have shown that antivitamin K anticoagulants and direct-acting anticoagulants do not increase the risk of progression to more severe stages. Antivitamin K consumption was associated with a lower risk of hospitalization and susceptibility to infection.
Collapse
Affiliation(s)
- Maruxa Zapata-Cachafeiro
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health, (CIBER en Epidemiología y Salud Pública-CIBERESP), Santiago de Compostela, Spain
- Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Ángela Prieto-Campo
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
| | - Manuel Portela-Romero
- Centro de Salud Concepción Arenal, Santiago de Compostela, A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Santiago de Compostela, Spain
| | - Eduardo Carracedo-Martínez
- Santiago de Compostela Health Area, Galician Health Service (Servizo Galego de Saúde-SERGAS), Santiago de Compostela, Spain
| | - Martina Lema-Oreiro
- Pharmaceutical Provision Management Service, Galician Health Service, Santiago de Compostela, Spain
| | - María Piñeiro-Lamas
- Consortium for Biomedical Research in Epidemiology and Public Health, (CIBER en Epidemiología y Salud Pública-CIBERESP), Santiago de Compostela, Spain
- Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Somnath Chaudhuri
- Research Group on Statistics, Econometrics and Health (GRECS), University of Girona, Girona, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Ángel Salgado-Barreira
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain.
- Consortium for Biomedical Research in Epidemiology and Public Health, (CIBER en Epidemiología y Salud Pública-CIBERESP), Santiago de Compostela, Spain.
- Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.
| | - Adolfo Figueiras
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health, (CIBER en Epidemiología y Salud Pública-CIBERESP), Santiago de Compostela, Spain
- Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
31
|
Abrashev H, Ananiev J, Georgieva E. Case Report: Spontaneous Left Inferior Epigastric Artery Injury in a COVID-19 Female Patient Undergoing Anticoagulation Therapy. J Clin Med 2023; 12:jcm12051842. [PMID: 36902629 PMCID: PMC10003174 DOI: 10.3390/jcm12051842] [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: 01/05/2023] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
Since the beginning of the pandemic, a recommendation was made for the use of anticoagulants in high-risk hospitalized patients. This therapeutic approach has positive and negative effects regarding the outcome of the disease. Anticoagulant therapy prevents thromboembolic events, but it can also lead to spontaneous hematoma formation, or be accompanied by massive active bleeding. We present a 63-year-old COVID-19-positive female patient with a massive retroperitoneal hematoma and spontaneous left inferior epigastric artery injury.
Collapse
Affiliation(s)
- Hristo Abrashev
- Department of Special Surgery, Orthopedics, and Traumatology, Clinic of Vascular Surgery, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria
| | - Julian Ananiev
- Department of General and Clinical Pathology, Forensic Medicine and Deontology and Dermatovenereology, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria
| | - Ekaterina Georgieva
- Department of General and Clinical Pathology, Forensic Medicine and Deontology and Dermatovenereology, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria
- Correspondence: ; Tel.: +359-878552006
| |
Collapse
|
32
|
Islahudin F, Low L, Saffian S. Factors of venous thromboembolism among COVID-19 patients. ASIAN JOURNAL OF PHARMACEUTICAL RESEARCH AND HEALTH CARE 2023. [DOI: 10.4103/ajprhc.ajprhc_13_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
33
|
Ammar LA, Nassar JE, Bitar F, Arabi M. COVID-19 in Cyanotic Congenital Heart Disease. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2023; 2023:5561159. [PMID: 37114013 PMCID: PMC10129433 DOI: 10.1155/2023/5561159] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023]
Abstract
Congenital heart disease (CHD) is the most prevalent congenital defect in newborn infants. Due to the various types of heart abnormalities, CHD can have a wide range of symptoms. Cardiac lesions comprise a range of different types and accordingly varying severities. It is highly helpful to classify CHD into cyanotic and acyanotic heart diseases. In this review, we are investigating the course of Coronavirus disease 2019 (COVID-19) in cyanotic CHD patients. The infection may directly or indirectly affect the heart by affecting the respiratory system and other organs. The effect on the heart that is pressure- or volume-overloaded in the context of CHD is theoretically more severe. Patients with CHD are at a higher risk of mortality from COVID-19 infection or suffering worse complications. While the anatomic complexity of CHD does not seem to predict the severity of infection, patients with worse physiological stages are more susceptible such as cyanosis and pulmonary hypertension. Patients with CHD exhibit continuous hypoxemia and have lower oxygen saturations because of a right-to-left shunt. Such individuals run the danger of rapidly deteriorating in the event of respiratory tract infections with inadequate oxygenation. Additionally, these patients have a higher risk of paradoxical embolism. Hence, critical care should be given to cyanotic heart disease patients with COVID-19 in comparison to acyanotic patients and this is through proper management, close observation, and adequate medical therapy.
Collapse
Affiliation(s)
- Lama A Ammar
- Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut 1107-2020, Lebanon
| | - Joseph E Nassar
- Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut 1107-2020, Lebanon
| | - Fadi Bitar
- Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut 1107-2020, Lebanon
| | - Mariam Arabi
- Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut 1107-2020, Lebanon
| |
Collapse
|
34
|
Chaudhary P, Alam S, Emon NU, Singh D, Janmeda P, Docea AO, Calina D, Sharifi-Rad J. COVID-19: An Overview of Virology, Mutations, Pathology, Epidemiology, Diagnosis, Preventions, and Treatments. ETHNOPHARMACOLOGY AND DRUG DISCOVERY FOR COVID-19: ANTI-SARS-COV-2 AGENTS FROM HERBAL MEDICINES AND NATURAL PRODUCTS 2023:1-22. [DOI: 10.1007/978-981-99-3664-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
35
|
Bruhn-Olszewska B, Davies H, Sarkisyan D, Juhas U, Rychlicka-Buniowska E, Wójcik M, Horbacz M, Jąkalski M, Olszewski P, Westholm JO, Smialowska A, Wierzba K, Torinsson Naluai Å, Jern N, Andersson LM, Järhult JD, Filipowicz N, Tiensuu Janson E, Rubertsson S, Lipcsey M, Gisslén M, Hultström M, Frithiof R, Dumanski JP. Loss of Y in leukocytes as a risk factor for critical COVID-19 in men. Genome Med 2022; 14:139. [PMID: 36514076 PMCID: PMC9747543 DOI: 10.1186/s13073-022-01144-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The COVID-19 pandemic, which has a prominent social and economic impact worldwide, shows a largely unexplained male bias for the severity and mortality of the disease. Loss of chromosome Y (LOY) is a risk factor candidate in COVID-19 due to its prior association with many chronic age-related diseases, and its impact on immune gene transcription. METHODS Publicly available scRNA-seq data of PBMC samples derived from male patients critically ill with COVID-19 were reanalyzed, and LOY status was added to the annotated cells. We further studied LOY in whole blood for 211 COVID-19 patients treated at intensive care units (ICU) from the first and second waves of the pandemic. Of these, 139 patients were subject to cell sorting for LOY analysis in granulocytes, low-density neutrophils (LDNs), monocytes, and PBMCs. RESULTS Reanalysis of available scRNA-seq data revealed LDNs and monocytes as the cell types most affected by LOY. Subsequently, DNA analysis indicated that 46%, 32%, and 29% of critically ill patients showed LOY above 5% cut-off in LDNs, granulocytes, and monocytes, respectively. Hence, the myeloid lineage that is crucial for the development of severe COVID-19 phenotype is affected by LOY. Moreover, LOY correlated with increasing WHO score (median difference 1.59%, 95% HDI 0.46% to 2.71%, p=0.025), death during ICU treatment (median difference 1.46%, 95% HDI 0.47% to 2.43%, p=0.0036), and history of vessel disease (median difference 2.16%, 95% HDI 0.74% to 3.7%, p=0.004), among other variables. In 16 recovered patients, sampled during ICU stay and 93-143 days later, LOY decreased significantly in whole blood and PBMCs. Furthermore, the number of LDNs at the recovery stage decreased dramatically (median difference 76.4 per 10,000 cell sorting events, 95% HDI 55.5 to 104, p=6e-11). CONCLUSIONS We present a link between LOY and an acute, life-threatening infectious disease. Furthermore, this study highlights LOY as the most prominent clonal mutation affecting the myeloid cell lineage during emergency myelopoiesis. The correlation between LOY level and COVID-19 severity might suggest that this mutation affects the functions of monocytes and neutrophils, which could have consequences for male innate immunity.
Collapse
Affiliation(s)
- Bożena Bruhn-Olszewska
- grid.8993.b0000 0004 1936 9457Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Hanna Davies
- grid.8993.b0000 0004 1936 9457Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Daniil Sarkisyan
- grid.8993.b0000 0004 1936 9457Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ulana Juhas
- grid.11451.300000 0001 0531 34263P-Medicine Laboratory, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Edyta Rychlicka-Buniowska
- grid.11451.300000 0001 0531 34263P-Medicine Laboratory, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Magdalena Wójcik
- grid.11451.300000 0001 0531 34263P-Medicine Laboratory, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Monika Horbacz
- grid.11451.300000 0001 0531 34263P-Medicine Laboratory, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Marcin Jąkalski
- grid.11451.300000 0001 0531 34263P-Medicine Laboratory, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Paweł Olszewski
- grid.11451.300000 0001 0531 34263P-Medicine Laboratory, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Jakub O. Westholm
- grid.10548.380000 0004 1936 9377National Bioinformatics Infrastructure Sweden, Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, Stockholm, Sweden
| | - Agata Smialowska
- grid.10548.380000 0004 1936 9377National Bioinformatics Infrastructure Sweden, Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, Stockholm, Sweden
| | - Karol Wierzba
- grid.11451.300000 0001 0531 3426Department and Clinic of Rheumatology, Clinical Immunology, Geriatrics and Internal Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Åsa Torinsson Naluai
- grid.8761.80000 0000 9919 9582Department of Laboratory Medicine, Institute of Biomedicine and Biobank Core Facility, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Niklas Jern
- grid.8761.80000 0000 9919 9582Department of Laboratory Medicine, Institute of Biomedicine and Biobank Core Facility, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Lars-Magnus Andersson
- grid.8761.80000 0000 9919 9582Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden ,grid.1649.a000000009445082XDepartment of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Josef D. Järhult
- grid.8993.b0000 0004 1936 9457Zoonosis Science Center, Department of Medical Sciences, Uppsala, Sweden, Uppsala University, Uppsala, Sweden
| | - Natalia Filipowicz
- grid.11451.300000 0001 0531 34263P-Medicine Laboratory, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Eva Tiensuu Janson
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Endocrine Oncology Unit, Uppsala University, Uppsala, Sweden
| | - Sten Rubertsson
- grid.8993.b0000 0004 1936 9457Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Miklós Lipcsey
- grid.8993.b0000 0004 1936 9457Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden ,grid.8993.b0000 0004 1936 9457Hedenstierna laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Magnus Gisslén
- grid.8761.80000 0000 9919 9582Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden ,grid.1649.a000000009445082XDepartment of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Michael Hultström
- grid.8993.b0000 0004 1936 9457Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden ,grid.8993.b0000 0004 1936 9457Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Robert Frithiof
- grid.8993.b0000 0004 1936 9457Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Jan P. Dumanski
- grid.8993.b0000 0004 1936 9457Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden ,grid.11451.300000 0001 0531 34263P-Medicine Laboratory, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| |
Collapse
|
36
|
Wei N, Xu Y, Wang H, Jia Q, Shou X, Zhang X, Zhang N, Li Y, Zhai H, Hu Y. Bibliometric and visual analysis of cardiovascular diseases and COVID-19 research. Front Public Health 2022; 10:1022810. [PMID: 36568760 PMCID: PMC9773213 DOI: 10.3389/fpubh.2022.1022810] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Background The global community has been affected by the coronavirus disease 2019 (COVID-19), which emerged in December 2019. Since then, many studies have been conducted on cardiovascular diseases (CVDs) and COVID-19. The aim of this study was to perform a bibliometric and visual analysis of the published relationship between CVDs and COVID-19. Methods 1,890 publications were retrieved from the Web of Science Core Collection database on January 5, 2022. Microsoft Office Excel and CiteSpace were then used to carry out scientometric analysis on the relevant literature according to seven aspects: document type, countries/regions, institutions, authors, journals, references, and keywords. Results The research on CVDs and COVID-19 is currently in a period of rapid development, with China, USA, England, and Italy leading the field. There is active cooperation between most countries and institutions. Harvard Medical School stands out among the many institutions not only for the largest number of publications, but also for their high quality. Banerjee A, Solomon SD and Narula J are three representative authors in this field. Frontiers in Cardiovascular Medicine was the journal with the highest number of published studies, and The Lancet was the most cited journal. Two documents with a high degree of significance in this field were identified. Popular research topics in this field are specific diseases, such as acute coronary syndrome and heart failure; pathogenesis related to ACE2, insulin resistance and pericyte; the specific therapeutic drug chloroquine; and clinical characteristics, physical activity, and mental health. ACE2 and NF-κB will be the focus of future research. Conclusions This study provides useful information for the research of CVDs and COVID-19, including potential collaborators, popular research topics, and a reference for more extensive and in-depth research in the future.
Collapse
Affiliation(s)
- Namin Wei
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Xu
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Huan Wang
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiulei Jia
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xintian Shou
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuesong Zhang
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nan Zhang
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ya'nan Li
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Huaqiang Zhai
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China,*Correspondence: Huaqiang Zhai
| | - Yuanhui Hu
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Yuanhui Hu
| |
Collapse
|
37
|
Aguilar-Andino D, Umaña AN, Alas-Pineda C, Santos FM, Gómez AC, Soto MM, Osorio AL. Clinical features, coagulation and inflammatory biomarkers associated with poor in-hospital outcomes in a Honduran population with RT-PCR confirmed COVID-19. THROMBOSIS UPDATE 2022; 9:100124. [PMID: 38620940 PMCID: PMC9529676 DOI: 10.1016/j.tru.2022.100124] [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: 01/16/2022] [Revised: 08/14/2022] [Accepted: 10/03/2022] [Indexed: 12/15/2022] Open
Abstract
Background SARS-COV-2, in most cases, only generates a mild acute respiratory disease. However, patients with severe disease show an exaggerated response of the immune system, creating a pro-inflammatory state, which could cause abnormalities in the coagulation system that increases mortality. Latin American countries, specially those with limited resources, have few studies about clinical features, coagulation and inflammatory biomarkers that could be useful at admission to assess poor outcomes. Objective The objective of this study is to describe the clinical features, coagulation, and inflammatory biomarkers, and identify risk factors at admission that are associated poor outcomes in Honduran population. Methods A cohort study was conducted. 210 patients were included, which 105 died during hospitalization due to COVID-19 and 105 were discharged alive, between September 2020 and January 2021. Clinical and laboratorial data was retrospectively collected. Results 57,6% of the population were male. The median age was 58 years. The median time between symptom onset and hospital admission was 6 days. D-dimer median was higher in the dead group compared with the alive group. Poor prognosis factors in the Cox multivariable model were male gender, age, symptom's duration, obesity and an elevated d dimer at admission. Conclusion In low-middle income countries, the assessment of these clinical and laboratory tools, especially in those with risk factors for prothrombotic states, could help clinicians to correctly stratify disease prognosis, establish a baseline to evaluate further evolution, and also predict outcomes, thus improving patient management.
Collapse
Affiliation(s)
- David Aguilar-Andino
- Departamento de Epidemiologia, Hospital General Dr. Mario Catarino Rivas, San Pedro Sula, Honduras
- Departamento de Medicina, Universidad Nacional Autónoma de Honduras, San Pedro Sula, Honduras
| | - Andrea N Umaña
- Facultad de Medicina y Cirugía, Universidad Católica de Honduras, Campus San Pedro y San Pablo, Honduras
| | - César Alas-Pineda
- Departamento de Epidemiologia, Hospital General Dr. Mario Catarino Rivas, San Pedro Sula, Honduras
- Facultad de Medicina y Cirugía, Universidad Católica de Honduras, Campus San Pedro y San Pablo, Honduras
| | - Freddy Medina Santos
- Departamento de Medicina Interna, Hospital General Dr. Mario Catarino Rivas, San Pedro Sula, Honduras
| | - Alejandro Cárcamo Gómez
- Departamento de Medicina Interna, Hospital General Dr. Mario Catarino Rivas, San Pedro Sula, Honduras
| | - Marco Molina Soto
- Departamento de Medicina Interna, Hospital General Dr. Mario Catarino Rivas, San Pedro Sula, Honduras
| | - Ana Liliam Osorio
- Departamento de Medicina Interna, Hospital General Dr. Mario Catarino Rivas, San Pedro Sula, Honduras
| |
Collapse
|
38
|
Ishigaki H, Yasui F, Nakayama M, Endo A, Yamamoto N, Yamaji K, Nguyen CT, Kitagawa Y, Sanada T, Honda T, Munakata T, Higa M, Toyama S, Kono R, Takagi A, Matsumoto Y, Koseki A, Hayashi K, Shiohara M, Ishii K, Saeki Y, Itoh Y, Kohara M. An attenuated vaccinia vaccine encoding the severe acute respiratory syndrome coronavirus-2 spike protein elicits broad and durable immune responses, and protects cynomolgus macaques and human angiotensin-converting enzyme 2 transgenic mice from severe acute respiratory syndrome coronavirus-2 and its variants. Front Microbiol 2022; 13:967019. [PMID: 36466631 PMCID: PMC9716133 DOI: 10.3389/fmicb.2022.967019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 10/14/2022] [Indexed: 08/27/2023] Open
Abstract
As long as the coronavirus disease-2019 (COVID-19) pandemic continues, new variants of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) with altered antigenicity will emerge. The development of vaccines that elicit robust, broad, and durable protection against SARS-CoV-2 variants is urgently required. We have developed a vaccine consisting of the attenuated vaccinia virus Dairen-I (DIs) strain platform carrying the SARS-CoV-2 S gene (rDIs-S). rDIs-S induced neutralizing antibody and T-lymphocyte responses in cynomolgus macaques and human angiotensin-converting enzyme 2 (hACE2) transgenic mice, and the mouse model showed broad protection against SARS-CoV-2 isolates ranging from the early-pandemic strain (WK-521) to the recent Omicron BA.1 variant (TY38-873). Using a tandem mass tag (TMT)-based quantitative proteomic analysis of lung homogenates from hACE2 transgenic mice, we found that, among mice subjected to challenge infection with WK-521, vaccination with rDIs-S prevented protein expression related to the severe pathogenic effects of SARS-CoV-2 infection (tissue destruction, inflammation, coagulation, fibrosis, and angiogenesis) and restored protein expression related to immune responses (antigen presentation and cellular response to stress). Furthermore, long-term studies in mice showed that vaccination with rDIs-S maintains S protein-specific antibody titers for at least 6 months after a first vaccination. Thus, rDIs-S appears to provide broad and durable protective immunity against SARS-CoV-2, including current variants such as Omicron BA.1 and possibly future variants.
Collapse
Affiliation(s)
- Hirohito Ishigaki
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Fumihiko Yasui
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Misako Nakayama
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Akinori Endo
- Protein Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Naoki Yamamoto
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kenzaburo Yamaji
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Cong Thanh Nguyen
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Yoshinori Kitagawa
- Division of Microbiology and Infectious Diseases, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Takahiro Sanada
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tomoko Honda
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tsubasa Munakata
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masahiko Higa
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Sakiko Toyama
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Risa Kono
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Asako Takagi
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yusuke Matsumoto
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Aya Koseki
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kaori Hayashi
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Masanori Shiohara
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Koji Ishii
- Department of Quality Assurance and Radiological Protection, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yasushi Saeki
- Protein Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yasushi Itoh
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
39
|
Practical Recommendations for Optimal Thromboprophylaxis in Patients with COVID-19: A Consensus Statement Based on Available Clinical Trials. J Clin Med 2022; 11:jcm11205997. [PMID: 36294316 PMCID: PMC9604499 DOI: 10.3390/jcm11205997] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/22/2022] [Accepted: 10/07/2022] [Indexed: 11/23/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been shown to be strongly associated with increased risk for venous thromboembolism events (VTE) mainly in the inpatient but also in the outpatient setting. Pharmacologic thromboprophylaxis has been shown to offer significant benefits in terms of reducing not only VTE events but also mortality, especially in acutely ill patients with COVID-19. Although the main source of evidence is derived from observational studies with several limitations, thromboprophylaxis is currently recommended for all hospitalized patients with acceptable bleeding risk by all national and international guidelines. Recently, high quality data from randomized controlled trials (RCTs) further support the role of thromboprophylaxis and provide insights into the optimal thromboprophylaxis strategy. The aim of this statement is to systematically review all the available evidence derived from RCTs regarding thromboprophylaxis strategies in patients with COVID-19 in different settings (either inpatient or outpatient) and provide evidence-based guidance to practical questions in everyday clinical practice. Clinical questions accompanied by practical recommendations are provided based on data derived from 20 RCTs that were identified and included in the present study. Overall, the main conclusions are: (i) thromboprophylaxis should be administered in all hospitalized patients with COVID-19, (ii) an optimal dose of inpatient thromboprophylaxis is dependent upon the severity of COVID-19, (iii) thromboprophylaxis should be administered on an individualized basis in post-discharge patients with COVID-19 with high thrombotic risk, and (iv) thromboprophylaxis should not be routinely administered in outpatients. Changes regarding the dominant SARS-CoV-2 variants, the wide immunization status (increasing rates of vaccination and reinfections), and the availability of antiviral therapies and monoclonal antibodies might affect the characteristics of patients with COVID-19; thus, future studies will inform us about the thrombotic risk and the optimal therapeutic strategies for these patients.
Collapse
|
40
|
Muacevic A, Adler JR. A Case of Deep Vein Thrombosis After Recovery From COVID-19 and Its Association With Elevated D-dimers. Cureus 2022; 14:e29859. [PMID: 36337799 PMCID: PMC9628276 DOI: 10.7759/cureus.29859] [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] [Accepted: 10/03/2022] [Indexed: 12/02/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) continues to be a devastating disease for the elderly population, especially in long-term care facilities, and it presents with varying clinical presentations. We have ample evidence that COVID-19 can predispose to deep vein thrombosis (DVT) and pulmonary embolism (PE) during an active infection. Still, very few cases of DVT have been reported after recovery from COVID-19. The imbalance of the coagulation cascade and the increased release of certain coagulation factors play an essential role in promoting hypercoagulability and vascular endothelial dysfunction. It leads to a rise in the level of fibrin degradation products, D-dimers, which can remain elevated for up to several weeks, even after recovery. It has been suggested that the risk of DVT occurring after recovering from COVID-19 remains high for up to three months. We report a case of a 77-year-old long-term care female resident at a nursing facility, ambulatory at baseline, who was noted to be COVID-19 positive upon routine facility-wide testing per department of health guidelines. She was asymptomatic during her 10-day quarantine period. D-dimer levels during routine labs were high (initial D-dimer level of 1.87 mg/L FEU {normal value: 0.19-0.52 mg/L FEU}), but the patient had no clinical signs and symptoms of DVT. Ultrasound of the bilateral legs was not performed due to low clinical suspicion. The patient received an enoxaparin DVT prophylaxis dose during the quarantine period. Follow-up D-dimer levels were done at frequent intervals after recovery, but D-dimer levels continued to remain elevated up till six weeks after her 10-day quarantine period ended. Based on previous experience with other long-term care residents who suffered from COVID-19, bilateral lower extremity ultrasound was performed, which showed bilateral DVT. Elevated D-dimer levels are a predictor of hypercoagulation complications in COVID-19. Patients with persistently elevated D-dimer levels after recovery from COVID-19 should be screened for thromboembolic complications, even if they are asymptomatic. DVT can occur up to three months post-recovery from COVID-19 infection.
Collapse
|
41
|
Grobbelaar LM, Kruger A, Venter C, Burger EM, Laubscher GJ, Maponga TG, Kotze MJ, Kwaan HC, Miller JB, Fulkerson D, Huff W, Chang E, Wiarda G, Bunch CM, Walsh MM, Raza S, Zamlut M, Moore HB, Moore EE, Neal MD, Kell DB, Pretorius E. Relative Hypercoagulopathy of the SARS-CoV-2 Beta and Delta Variants when Compared to the Less Severe Omicron Variants Is Related to TEG Parameters, the Extent of Fibrin Amyloid Microclots, and the Severity of Clinical Illness. Semin Thromb Hemost 2022; 48:858-868. [PMID: 36174604 DOI: 10.1055/s-0042-1756306] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Earlier variants of SARS-CoV-2 have been associated with plasma hypercoagulability (as judged by thromboelastography) and an extensive formation of fibrin amyloid microclots, which are considered to contribute to the pathology of the coronavirus 2019 disease (COVID-19). The newer Omicron variants appear to be far more transmissible, but less virulent, even when taking immunity acquired from previous infections or vaccination into account. We here show that while the clotting parameters associated with Omicron variants are significantly raised over those of healthy, matched controls, they are only raised to levels significantly lower than those seen with more severe variants such as beta and delta. We also observed that individuals infected with omicron variants manifested less extensive microclot formation in platelet-poor plasma compared with those harboring the more virulent variants. The measurement of clotting effects between the different variants acts as a kind of "internal control" that demonstrates the relationship between the extent of coagulopathies and the virulence of the variant of interest. This adds to the evidence that microclots may play an important role in reflecting the severity of symptoms observed in COVID-19.
Collapse
Affiliation(s)
- Lize M Grobbelaar
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Matieland, South Africa
| | - Arneaux Kruger
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Matieland, South Africa
| | - Chantelle Venter
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Matieland, South Africa
| | | | | | - Tongai G Maponga
- Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town, South Africa
| | - Maritha J Kotze
- Division of Chemical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University and National Health Laboratory Service, Tygerberg Hospital, Cape Town, South Africa
| | - Hau C Kwaan
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Joseph B Miller
- Departments of Emergency Medicine and Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Daniel Fulkerson
- Department of Neurosurgery, St. Joseph Regional Medical Center, Mishawaka, Indiana
| | - Wei Huff
- Department of Neurosurgery, St. Joseph Regional Medical Center, Mishawaka, Indiana
| | - Eric Chang
- Indiana University School of Medicine - South Bend, Notre Dame, Indiana
| | - Grant Wiarda
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, Indiana
| | - Connor M Bunch
- Departments of Emergency Medicine and Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Mark M Walsh
- Indiana University School of Medicine - South Bend, Notre Dame, Indiana.,Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, Indiana.,Department of Emergency Medicine, Saint Joseph Regional Medical Center, Mishawaka, Indiana
| | - Syed Raza
- Department of Critical Care Medicine, Saint Joseph Regional Medical Center, Mishawaka, Indiana
| | - Mahmud Zamlut
- Department of Critical Care Medicine, Saint Joseph Regional Medical Center, Mishawaka, Indiana
| | - Hunter B Moore
- Division of Transplant Surgery, Department of Surgery, Denver Health and University of Colorado Health Sciences Center, Denver, Colorado
| | - Ernest E Moore
- Department of Surgery, Ernest E. Moore Shock Trauma Center at Denver Health and University of Colorado Health Sciences Center, Denver, Colorado
| | - Matthew D Neal
- Pittsburgh Trauma Research Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Matieland, South Africa.,Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, England, United Kingdom.,The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Matieland, South Africa.,The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
42
|
Kubatka P, Mazurakova A, Koklesova L, Samec M, Sokol J, Samuel SM, Kudela E, Biringer K, Bugos O, Pec M, Link B, Adamkov M, Smejkal K, Büsselberg D, Golubnitschaja O. Antithrombotic and antiplatelet effects of plant-derived compounds: a great utility potential for primary, secondary, and tertiary care in the framework of 3P medicine. EPMA J 2022; 13:407-431. [PMID: 35990779 PMCID: PMC9376584 DOI: 10.1007/s13167-022-00293-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 12/29/2022]
Abstract
Thromboembolism is the third leading vascular disease, with a high annual incidence of 1 to 2 cases per 1000 individuals within the general population. The broader term venous thromboembolism generally refers to deep vein thrombosis, pulmonary embolism, and/or a combination of both. Therefore, thromboembolism can affect both - the central and peripheral veins. Arterial thromboembolism causes systemic ischemia by disturbing blood flow and oxygen supply to organs, tissues, and cells causing, therefore, apoptosis and/or necrosis in the affected tissues. Currently applied antithrombotic drugs used, e.g. to protect affected individuals against ischemic stroke, demonstrate significant limitations. For example, platelet inhibitors possess only moderate efficacy. On the other hand, thrombolytics and anticoagulants significantly increase hemorrhage. Contextually, new approaches are extensively under consideration to develop next-generation antithrombotics with improved efficacy and more personalized and targeted application. To this end, phytochemicals show potent antithrombotic efficacy demonstrated in numerous in vitro, ex vivo, and in vivo models as well as in clinical evaluations conducted on healthy individuals and persons at high risk of thrombotic events, such as pregnant women (primary care), cancer, and COVID-19-affected patients (secondary and tertiary care). Here, we hypothesized that specific antithrombotic and antiplatelet effects of plant-derived compounds might be of great clinical utility in primary, secondary, and tertiary care. To increase the efficacy, precise patient stratification based on predictive diagnostics is essential for targeted protection and treatments tailored to the person in the framework of 3P medicine. Contextually, this paper aims at critical review toward the involvement of specific classes of phytochemicals in antiplatelet and anticoagulation adapted to clinical needs. The paper exemplifies selected plant-derived drugs, plant extracts, and whole plant foods/herbs demonstrating their specific antithrombotic, antiplatelet, and fibrinolytic activities relevant for primary, secondary, and tertiary care. One of the examples considered is antithrombotic and antiplatelet protection specifically relevant for COVID-19-affected patient groups.
Collapse
Affiliation(s)
- Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Alena Mazurakova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Marek Samec
- Department of Pathological Physiology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Juraj Sokol
- Department of Hematology and Transfusion Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, 24144 Doha, Qatar
| | - Erik Kudela
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Kamil Biringer
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | | | - Martin Pec
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Barbara Link
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Karel Smejkal
- Department of Natural Drugs, Faculty of Pharmacy, Masaryk University, 61200 Brno, Czech Republic
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, 24144 Doha, Qatar
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| |
Collapse
|
43
|
Sedaghat F, Vadvala HV, Shan A, McMahon MT, Gawande RS. Incidence of Contrast-Associated Acute Kidney Injury in Renal-Competent COVID-19 Patients Undergoing Computed Chest Angiography. J Comput Assist Tomogr 2022; 46:701-706. [PMID: 35675687 PMCID: PMC9474721 DOI: 10.1097/rct.0000000000001337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/10/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE COVID-19 infection poses a significant risk of both renal injury and pulmonary embolism, producing a clinical challenge, as the criterion standard examination for pulmonary embolism, computed tomography angiography (CTA), requires the use of nephrotoxic iodinated contrast agents.Our investigation evaluated whether symptomatic COVID-19-positive patients without laboratory evidence of renal impairment are at increased risk for developing contrast-associated acute kidney injury (CA-AKI). METHOD All COVID-19-positive patients undergoing noncontrast chest computed tomography and CTA at an apex tertiary medical center between March 1 and December 10, 2020, were retrospectively evaluated. A total of 258 renal-competent (estimated glomerular filtration rate >30) patients with baseline and 48- to 72-hour postexamination creatinine measurements were identified and analyzed for incidence of acute kidney injury (AKI) meeting the criteria for CA-AKI. RESULTS Twenty-five of 191 patients undergoing CTA (13.1%) and 9 of the 67 undergoing noncontrast computed tomography (13.4%) experienced creatinine increases meeting the criteria for CA-AKI. Univariate and multivariate analyses accounting for known AKI risk factors revealed no correlation between iodinated contrast administration and the incidence AKI meeting the criteria for CA-AKI (univariable odds ratio, 0.97 [95% confidence interval, 0.43-2.20]; multivariable odds ratio, 0.97 [95% confidence interval, 0.40-2.36]). CONCLUSIONS Renal-competent COVID-19 patients undergoing chest CTA may not have an increased risk of AKI. Additional studies are needed to confirm this preliminary finding.
Collapse
Affiliation(s)
- Farzad Sedaghat
- From the Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD
| | - Harshna V. Vadvala
- From the Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD
| | - Alan Shan
- Cedars-Sinai Medical Center, Los Angeles, CA
| | - Michael T. McMahon
- From the Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD
| | - Rakhee S. Gawande
- From the Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD
| |
Collapse
|
44
|
Tekle E, Gelaw Y, Dagnew M, Gelaw A, Negash M, Kassa E, Bizuneh S, Wudineh D, Asrie F. Risk stratification and prognostic value of prothrombin time and activated partial thromboplastin time among COVID-19 patients. PLoS One 2022; 17:e0272216. [PMID: 35951632 PMCID: PMC9371343 DOI: 10.1371/journal.pone.0272216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/15/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND COVID-19 is a viral disease caused by a new strain of corona virus. Currently, prognosis and risk stratification of COVID-19 patients is done by the disease's clinical presentation. Therefore, identifying laboratory biomarkers for disease prognosis and risk stratification of COVID-19 patients is critical for prompt treatment. Therefore, the main objective of this study was to assess the risk stratification and prognostic value of basic coagulation parameters and factors associated with disease severity among COVID-19 patients at the Tibebe Ghion Specialized Hospital, COVID-19 treatment center, Northwest Ethiopia. METHODS A follow-up study was conducted among conveniently recruited COVID-19 patients attended from March to June 2021. Socio-demographic and clinical data were collected using a structured questionnaire and checklist, respectively. Prothrombin time (PT) and activated partial thromboplastin time (APTT) were analyzed by the HUMACLOT DUE PLUS® machine. Descriptive statistics were used to summarize the socio-demographic and clinical characteristics of study participants. Kruskal Wallis tests were used to compare the difference between parametric and non-parametric continuous variables, respectively. The area under the receiver operating characteristic curve (AUC) was used to evaluate the value of PT and APTT in the risk stratification and disease prognosis of COVID-19 patients. Ordinal logistic regression was used to identify the factors associated with disease severity and prognosis. A P-value < 0.05 was defined as statistically significant for all results. RESULT Baseline PT at a cut-off value ≥ 16.25 seconds differentiated severe COVID-19 patients from mild and moderate patients (AUC: 0.89, 95% CI: 0.83-0.95). PT also differentiated mild COVID-19 patients from moderate and severe patients at a cut-off value ≤ 15.35 seconds (AUC: 0.90, 95% CI: 0.84-0.96). Moreover, alcohol drinkers were a 3.52 times more likely chance of having severe disease than non-drinkers (95% CI: 1.41-8.81). A one-year increment in age also increased the odds of disease severity by 6% (95% CI: 3-9%). An increment of ≥ 0.65 seconds from the baseline PT predicted poor prognosis (AUC: 0.93, 0.87-0.99). CONCLUSIONS AND RECOMMENDATIONS Prolonged baseline PT was observed in severe COVID-19 patients. Prolonged baseline PT was also predicted to worsen prognosis. An increase from the baseline PT was associated with worsen prognosis. Therefore, PT can be used as a risk stratification and prognostic marker in COVID-19 patients.
Collapse
Affiliation(s)
- Esayas Tekle
- Department of Medical Laboratory Sciences, Institute of Health Sciences, Wollega University, Nekemte, Ethiopia
| | - Yemataw Gelaw
- Department of Hematology and Immunohematology, College of Medicine and Health Sciences, School of Biomedical and Laboratory Sciences, University of Gondar, Gondar, Ethiopia
| | - Mulat Dagnew
- Department of Medical Microbiology, College of Medicine and Health Sciences, School of Biomedical and Laboratory Sciences, University of Gondar, Gondar, Ethiopia
| | - Aschalew Gelaw
- Department of Medical Microbiology, College of Medicine and Health Sciences, School of Biomedical and Laboratory Sciences, University of Gondar, Gondar, Ethiopia
| | - Markos Negash
- Department of Immunology and Molecular Biology, College of Medicine and Health Sciences, School of Biomedical and Laboratory Sciences, University of Gondar, Gondar, Ethiopia
| | - Eyuel Kassa
- College of Medicine and Health Sciences, University of Gondar Comprehensive Specialized Hospital Laboratory, University of Gondar, Gondar, Ethiopia
| | - Segenet Bizuneh
- College of Medicine and Health Sciences, School of Medicine, University of Gondar, Gondar, Ethiopia
| | - Dessalew Wudineh
- Department of Medical Laboratory Sciences, Institute of Health Sciences, Mizan Tepi University, Mizan Tepi, Ethiopia
| | - Fikir Asrie
- Department of Hematology and Immunohematology, College of Medicine and Health Sciences, School of Biomedical and Laboratory Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
45
|
McDaniel CG, Commander SJ, DeLaura I, Cantrell S, Leraas HJ, Moore CB, Reed CR, Pahl KS, Tracy ET. Coagulation Abnormalities and Clinical Complications in Children With SARS-CoV-2: A Systematic Review of 48,322 Patients. J Pediatr Hematol Oncol 2022; 44:323-335. [PMID: 34862349 DOI: 10.1097/mph.0000000000002321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/07/2021] [Indexed: 02/03/2023]
Abstract
Given the limited information on the coagulation abnormalities of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in pediatric patients, we designed a systematic review to evaluate this topic. A comprehensive literature search was conducted for "SARS-CoV-2," "coagulopathy," and "pediatrics." Two authors independently screened the articles that the search returned for bleeding, thrombosis, anticoagulant and/or antiplatelet usage, and abnormal laboratory markers in pediatric patients with SARS-CoV-2, and the authors then extracted the relevant data. One hundred twenty-six publications were included. Thirty-four (27%) studies reported thrombotic complications in 504 patients. Thirty-one (25%) studies reported bleeding complications in 410 patients. Ninety-eight (78%) studies reported abnormal laboratory values in 6580 patients. Finally, 56 (44%) studies reported anticoagulant and/or antiplatelet usage in 3124 patients. The variety of laboratory abnormalities and coagulation complications associated with SARS-CoV-2 presented in this review highlights the complexity and variability of the disease presentation in infants and children.
Collapse
Affiliation(s)
| | | | | | - Sarah Cantrell
- Duke University School of Medicine
- Duke University Medical Center Library and Archives, Durham, NC
| | | | | | | | - Kristy S Pahl
- Division of Pediatric Hematology-Oncology
- Department of Pediatrics
| | - Elisabeth T Tracy
- Department of Surgery
- Division of Pediatric Surgery, Duke University Medical Center
| |
Collapse
|
46
|
COVID-19 tanısı konmuş gebe kadınlarda proinflamatuar IL-6 ve antiinflamatuar IL-10 sitokinlerinin etkileri. ANADOLU KLINIĞI TIP BILIMLERI DERGISI 2022. [DOI: 10.21673/anadoluklin.1129488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
ÖZ
Amaç: Çalışmamızda COVID-19 pozitif gebelerde serum interlökin-6 ve interlökin-10 düzeyleri ile hastalığın seyri arasındaki ilişkinin araştırılması amaçlandı.
Yöntemler: Çalışmamızda 28 üçüncü trimester COVID-19 pozitif gebe ve 30 üçüncü trimester sağlıklı gebede serum IL-6 ve IL-10 düzeyleri ölçüldü. COVID-19 pozitif vakalar, taşıyıcı veya hasta olarak sınıflandırıldı. Çalışma grubundaki gebelerden 13'ü asemptomatik iken veya hafif hastalık nedeniyle takip edilirken; 7'si yoğun bakımda (YBÜ) olmak üzere toplam 15 hamile kadın hastaneye kaldırılarak tedavi altına alındı. COVID-19 pozitif gebe kadınların IL-6 ve IL-10 testleri ilk uygulama sırasında çalışıldı.
Bulgular: COVID-19'lu 7 (%25) hastanın yoğun bakım ünitesine kabul edilmesi gerekti. COVID-19 negatif gebelerde IL-6 düzeyi, COVID-19 taşıyıcısı ve hasta gebelere göre anlamlı derecede düşük bulundu (p=0,01). COVID-19 negatif olan gebelerde IL-10 düzeyi, COVID-19 taşıyıcısı olan gebelere (p=0,002) ve hastalara (p=0,002) göre anlamlı derecede yüksek bulundu.
Sonuç: Şüpheli veya doğrulanmış bir COVID-19 teşhisi ile başvuran hamile kadınlarda olumsuz sonuç riskini en aza indirmek için IL-6 ve IL-10 sitokin düzeylerinin yakından izlenmesi önerilir. Bu şekilde hamile kadınlarda orta-hafif COVID-19'u şiddetli COVID-19'dan ayırt etmek mümkün olabilir.
Collapse
|
47
|
Johnson SE, Pai E, Voroba A, Chen NW, Bahl A. Examining D-dimer and Empiric Anti-coagulation in COVID-19-Related Thrombosis. Cureus 2022; 14:e26883. [PMID: 35978762 PMCID: PMC9375952 DOI: 10.7759/cureus.26883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Thrombosis is thought to occur frequently in the setting of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We aimed to elucidate the relationship between macro/microvascular thrombosis, D-dimer levels, and empiric anticoagulation in coronavirus disease 2019 (COVID-19). Methods This was an exploratory prospective, single-site, observational study. Adult emergency department patients with COVID-19 requiring hospitalization received a point-of-care lower extremity venous duplex ultrasound. The primary endpoint was thromboembolism and associated D-dimer level. Secondary endpoints included rates of micro and macro thrombotic complications as well as empiric anticoagulant use. Results Between January 13th and April 12th 2021, 52 patients were enrolled. Median D-dimer at presentation was 650 ng/mL (range 250-10,000 ng/mL) among patients with negative duplex studies. During hospitalization, 18 patients underwent 20 additional studies assessing for venous thromboembolism (VTE). These studies yielded one deep vein thrombosis (DVT) diagnosis. Among patients with negative studies median D-dimer was 1,246 ng/mL (range 329-10,000 ng/mL). Two patients experienced microvascular complications. Seven patients were started on empiric full dose anticoagulation. Conclusion While VTE remains a major concern amongst patients with COVID-19, the normal D-dimer cut off of >500 ng/mL likely should not be used to initiate further VTE workup. Additionally, moderately elevated D-dimer did not correlate strongly with microvascular complications and may not be relevant in the decision to initiate empiric anticoagulation.
Collapse
|
48
|
El‐Qushayri AE, Reda A, Dahy A, Azzam AY, Ghozy S. The impact of COVID 19 on the outcomes of thrombectomy in stroke patients: A systematic review and meta-analysis. Rev Med Virol 2022; 33:e2379. [PMID: 35833712 PMCID: PMC9349746 DOI: 10.1002/rmv.2379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/22/2022] [Accepted: 07/05/2022] [Indexed: 01/28/2023]
Abstract
We aimed to conduct the current meta-analysis to provide better insight into the efficacy of mechanical thrombectomy (MT) in managing COVID-19 patients suffering from a stroke. An electronic search was conducted through eight databases for collecting the current evidence about the efficacy of MT in stroke patients with COVID-19 until 18 December 2021. The results were reported as the pooled prevalence rates and the odds ratios (ORs), with their corresponding 95% confidence intervals (CI). Out of 648 records, we included nine studies. The prevalence of stroke patients with COVID-19 who received MT treatment was with TICI ≥2b 79% (95%CI: 73-85), symptomatic intracranial haemorrhage 6% (95%CI: 3-11), parenchymal haematoma type 1, 11.1% (95%CI: 5-23), and mortality 29% (95%CI: 24-35). On further comparison of MT procedure between stroke patients with COVID 19 to those without COVID-19, we found no significant difference in terms of TICI ≥2b score (OR: 0.85; 95%CI: 0.03-23; p = 0.9). However, we found that stroke patients with COVID-19 had a significantly higher mortality rate than stroke patients without COVID-19 after MT procedure (OR: 2.99; 95%CI: 2.01-4.45; p < 0.001). Stroke patients with COVID-19 can be safely and effectively treated with MT, with comparable reperfusion and complication rates to those without the disease.
Collapse
Affiliation(s)
| | | | | | | | - Sherief Ghozy
- Department of NeuroradiologyMayo ClinicRochesterMinnesotaUSA,Nuffield Department of Primary Care Health Sciences and Department for Continuing Education (EBHC program)Oxford UniversityOxfordUK
| |
Collapse
|
49
|
Álvarez‐Troncoso J, Ramos‐Ruperto L, Fernández‐Cidón P, Trigo‐Esteban E, Tung‐Chen Y, Busca‐Arenzana C, Quintana‐Díaz M, Buño‐Soto A, Arnalich‐Fernández F, Fernández‐Capitán C. Screening Protocol and Prevalence of Venous Thromboembolic Disease in Hospitalized Patients With COVID-19. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2022; 41:1689-1698. [PMID: 34694032 PMCID: PMC8661624 DOI: 10.1002/jum.15850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 09/04/2021] [Accepted: 09/20/2021] [Indexed: 05/18/2023]
Abstract
BACKGROUND SARS-CoV-2 disease (COVID-19) induces endothelial damage and sustained hypoxia and facilitates immobilization as factors of hypercoagulability. OBJECTIVES The objective of our study was to assess the prevalence of venous thromboembolic disease (VTD) in COVID-19 patients and the usefulness of VTD screening based on age-adjusted D-dimer and point-of-care ultrasound (POCUS). PATIENTS/METHODS We conducted a single cohort, prospective observational study in 102 consecutive hospitalized patients. RESULTS A total of 102 POCUS and 39 pulmonary computed tomography angiography (PCTA) were performed diagnosing 27 VTD (26.5%): 17 deep vein thrombosis (DVT) (16.6% positive POCUS) and 18 pulmonary embolism (PE) (46.2% positive PCTA). COVID-19 patients with VTD were older (P < .030), had higher D-dimer (P < .001), higher International Society on Thrombosis and Hemostasis score (P < .001), and higher mortality (P = .025). However, there were no differences in inflammatory laboratory parameters neither in the cytokine storm syndrome (CSS) development. The ROC curve for D-dimer showed an AUC of 0.91. We have evidenced that patients with D-dimer between 2000 and 6000 ng/mL could benefit from a screening strategy with POCUS given the high sensitivity and specificity of the test. Furthermore, patients with D-dimer ≥6000 ng/mL should undergo POCUS and PCTA to rule out DVT and PE, respectively. CONCLUSIONS In our cohort, 26.5% of the patients presented VTD. Screening strategy based on age-adjusted D-dimer and POCUS proved high sensitivity and specificity. Future trials focused on screening strategies are necessary to early detect the presence of DVT and PE and determine thromboprophylaxis strategies in patients with COVID-19.
Collapse
Affiliation(s)
| | | | | | | | - Yale Tung‐Chen
- Department of Emergency MedicineHospital Universitario La PazMadridSpain
| | | | | | - Antonio Buño‐Soto
- Department of Clinical AnalysisHospital Universitario La PazMadridSpain
| | | | | |
Collapse
|
50
|
Khalighi E, Marzban-Rad S, Taheri HR. Acute Progressive retro-peritoneal hematoma in COVID19 patients with sub cutaneous ecchymosis. Ann Med Surg (Lond) 2022; 79:104107. [PMID: 35784949 PMCID: PMC9238019 DOI: 10.1016/j.amsu.2022.104107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/26/2022] [Accepted: 06/26/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction and importance: COVID19 is a multifunction disease where hematological disorders are reported. Coagulopathy is seen in these patients, indicated by thromboembolic events. Case presentation We present case of 11 COVID19 who were presented with localized skin ecchymosis lesions in different areas of the body and retroperitoneal hematoma in the posterior wall of the abdomen and chest. Clinical discussion Increased INR and bleeding and ecchymosis in Corona patients require discontinuation of anticoagulants and, FFP, essential and tri amino injections are needed. Conclusions Diagnosis and management of hematoma is important to avoid fatality.
Global upsurge in COVID19 cases has been marked with systemic manifestations. We present case of 11 COVID19 who were presented with localized skin ecchymosis lesions. We present COVID19 patients who were treated with anticoagulants and were seen to be presented with retroperitoneal hematoma.
Collapse
Affiliation(s)
- Ebrahim Khalighi
- Department of Anesthesiology, School of Allied Medical Sciences, Shahid Mostafa Khomaeini Hospital, Ilam University of Medical sciences, Ilam, Iran
| | - Saeid Marzban-Rad
- Department of Surgery, Imam-Reza Hospital, Aja University of Medical Sciences, Tehran, Iran
- Corresponding author.
| | - Hamid Reza Taheri
- Department of Surgery, School of Medicine, Shahed University, Tehran, Iran
- Corresponding author.
| |
Collapse
|