51
|
Kiraz A, Sezer O, Alemdar A, Canbek S, Duman N, Bisgin A, Cora T, Ruhi HI, Ergoren MC, Geçkinli BB, Sag SO, Gözden HE, Oz O, Altıntaş ZM, Yalcıntepe S, Keskin A, Tak AY, Paskal ŞA, Yürekli UF, Demirtas M, Evren EU, Hanta A, Başdemirci M, Suer K, Balta B, Kocak N, Karabulut HG, Cobanogulları H, Ateş EA, Bozdoğan ST, Eker D, Ekinci S, Nergiz S, Tuncalı T, Yagbasan S, Alavanda C, Kutlay NY, Evren H, Erdoğan M, Altıner S, Sanlidag T, Gonen GA, Vicdan A, Eras N, Eker HK, Balasar O, Tuncel G, Dundar M, Gurkan H, Temel SG. Contribution of genotypes in Prothrombin and Factor V Leiden to COVID-19 and disease severity in patients at high risk for hereditary thrombophilia. J Med Virol 2023; 95:e28457. [PMID: 36597901 DOI: 10.1002/jmv.28457] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/23/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023]
Abstract
Thrombotic and microangiopathic effects have been reported in COVID-19 patients. This study examined the contribution of the hereditary thrombophilia factors Prothrombin (FII) and Factor V Leiden (FVL) genotypes to the severity of COVID-19 disease and the development of thrombosis. This study investigated FII and FVL alleles in a cohort of 9508 patients (2606 male and 6902 female) with thrombophilia. It was observed that 930 of these patients had been infected by SARS-CoV-2 causing COVID-19. The demographic characteristics of the patients and their COVID-19 medical history were recorded. Detailed clinical manifestations were analyzed in a group of cases (n = 4092). This subgroup was age and gender-matched. FII and FVL frequency data of healthy populations without thrombophilia risk were obtained from Bursa Uludag University Medical Genetic Department's Exome Databank. The ratio of males (31.08%; 27.01%) and the mean age (36.85 ± 15.20; 33.89 ± 14.14) were higher among COVID-19 patients compared to non-COVID-19 patients. The prevalence of FVL and computerized tomography (CT) positivity in COVID-19 patients was statistically significant in the thrombotic subgroup (p < 0.05). FVL prevalence, CT positivity rate, history of thrombosis, and pulmonary thromboembolism complication were found to be higher in deceased COVID-19 patients (p < 0.05). Disease severity was mainly affected by FVL and not related to genotypes at the Prothrombin mutations. Overall, disease severity and development of thrombosis in COVID-19 are mainly affected by the variation within the FVL gene. Possible FVL mutation should be investigated in COVID-19 patients and appropriate treatment should be started earlier in FVL-positive patients.
Collapse
Affiliation(s)
- Aslıhan Kiraz
- Kayseri City Training and Research Hospital, Genetic Diseases Evaluation Center, Kayseri, Turkey
| | - Ozlem Sezer
- Samsun Training and Research Hospital, Genetic Diseases Evaluation Center, Samsun, Turkey
| | - Adem Alemdar
- Department of Translational Medicine, Institute of Health Sciences, Bursa Uludag University, Bursa, Turkey
| | - Sezin Canbek
- Umraniye Training and Research Hospital, Genetic Diseases Evaluation Center, Health Sciences University, Istanbul, Turkey
| | - Nilgun Duman
- Department of Medical Genetics, Dragos Hospital, Bezmialem Vakıf University, Istanbul, Turkey
| | - Atıl Bisgin
- Medical Genetics Department of Medical Faculty, AGENTEM (Adana Genetic Diseases Diagnosis and Treatment Center), Cukurova University, Adana, Turkey
| | - Tulin Cora
- Department of Medical Genetics, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Hatice Ilgın Ruhi
- Department of Medical Genetics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Mahmut Cerkez Ergoren
- Department of Medical Genetics, Faculty of Medicine, Near East University, Nicosia, Cyprus
| | - Bilgen Bilge Geçkinli
- Department of Medical Genetics, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Sebnem Ozemri Sag
- Department of Medical Genetics, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Hilmi Erdem Gözden
- Department of Translational Medicine, Institute of Health Sciences, Bursa Uludag University, Bursa, Turkey.,Department of Haematology, Bursa Yuksek Ihtısas Training and Research Hospital, Health Sciences University, Bursa, Turkey
| | - Ozlem Oz
- Department of Medical Genetics, Faculty of Medicine, Harran University, Sanlıurfa, Turkey
| | - Zuhal Mert Altıntaş
- Department of Medical Genetics, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Sinem Yalcıntepe
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Adem Keskin
- Department of Biochemistry, Institute of Health Sciences, Adnan Menderes University, Aydın, Turkey
| | - Ayşegül Yabacı Tak
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Bezmialem Vakıf University, Istanbul, Turkey
| | - Şeyma Aktaş Paskal
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Uğur Fahri Yürekli
- Department of Medical Biochemistry, Sanlıurfa Mehmet Akif İnan Health Application and Research Center, Health Sciences University, Sanlıurfa, Turkey
| | | | - Emine Unal Evren
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, University of Kyrenia, Kyrenia, Cyprus
| | - Abdullah Hanta
- Cukurova University AGENTEM (Adana Genetic Diseases Diagnosis and Treatment Center), Adana, Turkey
| | - Müşerref Başdemirci
- Konya Training and Research Hospital, Genetic Diseases Evaluation Center, Health Sciences University, Konya, Turkey
| | - Kaya Suer
- Department of Infectious Diseases and Clinicai Microbiology, Faculty of Medicine, Near East University, Nicosia, Cyprus
| | - Burhan Balta
- Kayseri City Training and Research Hospital, Genetic Diseases Evaluation Center, Kayseri, Turkey
| | - Nadir Kocak
- Department of Medical Genetics, Faculty of Medicine, Selcuk University, Konya, Turkey
| | | | | | - Esra Arslan Ateş
- Department of Medical Genetics, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Sevcan Tuğ Bozdoğan
- Medical Genetics Department of Medical Faculty, AGENTEM (Adana Genetic Diseases Diagnosis and Treatment Center), Cukurova University, Adana, Turkey
| | - Damla Eker
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Sadiye Ekinci
- Department of Medical Genetics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Süleyman Nergiz
- Department of Medical Genetics, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Timur Tuncalı
- Department of Medical Genetics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Serap Yagbasan
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Ceren Alavanda
- Department of Medical Genetics, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Nuket Yurur Kutlay
- Department of Medical Genetics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Hakan Evren
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, University of Kyrenia, Kyrenia, Cyprus
| | - Murat Erdoğan
- Kayseri City Training and Research Hospital, Genetic Diseases Evaluation Center, Kayseri, Turkey
| | - Sule Altıner
- Department of Medical Genetics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | | | - Gizem Akıncı Gonen
- Kayseri City Training and Research Hospital, Genetic Diseases Evaluation Center, Kayseri, Turkey
| | - Arzu Vicdan
- Department of Medical Genetics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Nazan Eras
- Department of Medical Genetics, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Hatice Koçak Eker
- Konya Training and Research Hospital, Genetic Diseases Evaluation Center, Health Sciences University, Konya, Turkey
| | - Ozgür Balasar
- Konya Training and Research Hospital, Genetic Diseases Evaluation Center, Health Sciences University, Konya, Turkey
| | - Gulten Tuncel
- DESAM Institute, Near East University, Nicosia, Cyprus
| | - Munis Dundar
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Hakan Gurkan
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Sehime Gulsun Temel
- Department of Translational Medicine, Institute of Health Sciences, Bursa Uludag University, Bursa, Turkey.,Department of Medical Genetics, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey.,Department of Medical Genetics, Health Sciences Institute, Baskent University, Ankara, Turkey
| |
Collapse
|
52
|
Blaskovich MAT, Verderosa AD. Use of Antiviral Agents and other Therapies for COVID-19. Semin Respir Crit Care Med 2023; 44:118-129. [PMID: 36646090 DOI: 10.1055/s-0042-1758837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic led to a remarkably rapid development of a range of effective prophylactic vaccines, including new technologies that had not previously been approved for human use. In contrast, the development of new small molecule antiviral therapeutics has taken years to produce the first approved drugs specifically targeting severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), with the intervening years filled with attempts to repurpose existing drugs and the development of biological therapeutics. This review will discuss the reasons behind this variation in timescale and provide a survey of the many new treatments that are progressing through the clinical pipeline.
Collapse
Affiliation(s)
- Mark A T Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Anthony D Verderosa
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
53
|
Effect of troponin I and coagulation parameters on mortality in COVID-19 patients. MARMARA MEDICAL JOURNAL 2023. [DOI: 10.5472/marumj.1235703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Objective: Our aim is to determine the levels of troponin-I and some coagulation markers (D-dimer, fibrinogen and International Normalized Ratio (INR)) in coronavirus disease 2019 (COVID-19) patients and to investigate the effects of these markers on mortality.
Patients and Methods: It is planned as a descriptive, cross-sectional and analytical study. The study was conducted by retrospectively scanning the files of COVID-19 patients who applied to Inonu University Turgut Ozal Medical Center between 01.03.2020 and 31.12.2020. Levels of cardiac troponin I markers and coagulation parameters (D-dimer, fibrinogen and INR) were detected.
Results: The results of a total of 1858 patients were obtained. One thousand, three hundred and twenty-six patients with only troponin I and D-dimer results (Group 1), 606 patients with only troponin I and fibrinogen results (Group 2), and 1308 patients with only troponin I and INR results (Group 3) were included. Troponin I levels were significantly higher in all patients who died. 96.6% of the patients with high D-dimer levels died in Group 1, 85.5% of the patients with high fibrinogen levels died in Group 2 and 77.3 % of the patients with high INR levels died in Group 3.
Conclusion: Measurements of troponin-I and coagulation markers such as D-dimer, fibrinogen and INR can help predict clinical severity and mortality in COVID-19 patients.
Collapse
|
54
|
Rando HM, Lordan R, Kolla L, Sell E, Lee AJ, Wellhausen N, Naik A, Kamil JP. The Coming of Age of Nucleic Acid Vaccines during COVID-19. ARXIV 2023:arXiv:2210.07247v2. [PMID: 36263086 PMCID: PMC9580386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In the 21st century, several emergent viruses have posed a global threat. Each pathogen has emphasized the value of rapid and scalable vaccine development programs. The ongoing SARS-CoV-2 pandemic has made the importance of such efforts especially clear. New biotechnological advances in vaccinology allow for recent advances that provide only the nucleic acid building blocks of an antigen, eliminating many safety concerns. During the COVID-19 pandemic, these DNA and RNA vaccines have facilitated the development and deployment of vaccines at an unprecedented pace. This success was attributable at least in part to broader shifts in scientific research relative to prior epidemics; the genome of SARS-CoV-2 was available as early as January 2020, facilitating global efforts in the development of DNA and RNA vaccines within two weeks of the international community becoming aware of the new viral threat. Additionally, these technologies that were previously only theoretical are not only safe but also highly efficacious. Although historically a slow process, the rapid development of vaccines during the COVID-19 crisis reveals a major shift in vaccine technologies. Here, we provide historical context for the emergence of these paradigm-shifting vaccines. We describe several DNA and RNA vaccines and in terms of their efficacy, safety, and approval status. We also discuss patterns in worldwide distribution. The advances made since early 2020 provide an exceptional illustration of how rapidly vaccine development technology has advanced in the last two decades in particular and suggest a new era in vaccines against emerging pathogens.
Collapse
Affiliation(s)
- Halie M Rando
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America; Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America; Center for Health AI, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America; Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5158, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA
| | - Likhitha Kolla
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elizabeth Sell
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Alexandra J Lee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nils Wellhausen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Amruta Naik
- Children's Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Jeremy P Kamil
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
55
|
Varlas VN, Borș RG, Plotogea M, Iordache M, Mehedințu C, Cîrstoiu MM. Thromboprophylaxis in Pregnant Women with COVID-19: An Unsolved Issue. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1949. [PMID: 36767320 PMCID: PMC9915713 DOI: 10.3390/ijerph20031949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 06/18/2023]
Abstract
SARS-CoV-2 infection in pregnant women is of growing interest due to controversy over the use of antiplatelet and/or anticoagulant drugs during pregnancy and postpartum. Pregnant women are susceptible to develop severe forms of viral infections due to pregnancy-related immune alterations, changes in lung functions, and hypercoagulability. The association of pregnancy with SARS-CoV-2 infection can cause an increased incidence of thrombotic complications, especially in the case of patients with some genetic variants that favor inflammation and thrombosis. Compared to the general population, pregnant women may be at increased risk of thrombotic complications related to COVID-19. The lack of extensive clinical trials on thromboprophylaxis and extrapolating data from non-pregnant patients lead to major discrepancies in treating pregnant women with COVID-19. Currently, a multidisciplinary team should determine the dose and duration of prophylactic anticoagulant therapy for these patients, depending on the disease severity, the course of pregnancy, and the estimated due date. This narrative review aims to evaluate the protective effect of thromboprophylaxis in pregnant women with COVID-19. It is unknown at this time whether antiplatelet or anticoagulant therapy initiated at the beginning of pregnancy for various diseases (preeclampsia, intrauterine growth restriction, thrombophilia) offers a degree of protection. The optimal scheme for thromboprophylaxis in pregnant women with COVID-19 must be carefully established through an individualized decision concerning gestational age and the severity of the infection.
Collapse
Affiliation(s)
- Valentin Nicolae Varlas
- Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, 011171 Bucharest, Romania
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu St., 050451 Bucharest, Romania
| | - Roxana Georgiana Borș
- Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, 011171 Bucharest, Romania
| | - Mihaela Plotogea
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu St., 050451 Bucharest, Romania
- Department of Obstetrics and Gynecology, Nicolae Malaxa Clinical Hospital Bucharest, 022441 Bucharest, Romania
| | - Madalina Iordache
- Doctoral School, “Carol Davila”, University of Medicine and Pharmacy, 4192910 Bucharest, Romania
| | - Claudia Mehedințu
- Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, 011171 Bucharest, Romania
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu St., 050451 Bucharest, Romania
| | - Monica Mihaela Cîrstoiu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu St., 050451 Bucharest, Romania
- Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, 050098 Bucharest, Romania
| |
Collapse
|
56
|
Fang Y, Lin S, Dou Q, Gui J, Li W, Tan H, Wang Y, Zeng J, Khan A, Wei DQ. Network pharmacology- and molecular simulation-based exploration of therapeutic targets and mechanisms of heparin for the treatment of sepsis/COVID-19. J Biomol Struct Dyn 2023; 41:12586-12598. [PMID: 36661370 DOI: 10.1080/07391102.2023.2167114] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/05/2023] [Indexed: 01/21/2023]
Abstract
Critically infected patients with COVID-19 (coronavirus disease 2019) are prone to develop sepsis-related coagulopathy as a result of a robust immune response. The mechanism underlying the relationship between sepsis and COVID-19 is largely unknown. LMWH (low molecular weight heparin) exhibits both anti-inflammatory and anti-coagulating properties that result in a better prognosis of severely ill patients with COVID-19 co-associated with sepsis-induced coagulopathy or with a higher D-dimer value. Heparin-associated molecular targets and their mechanism of action in sepsis/COVID-19 are not well understood. In this work, we characterize the pharmacological targets, biological functions and therapeutic actions of heparin in sepsis/COVID-19 from the perspective of network pharmacology. A total of 38 potential targets for heparin action against sepsis/COVID-19 and 8 core pharmacological targets were identified, including IL6, KNG1, CXCL8, ALB, VEGFA, F2, IL10 and TNF. Moreover, enrichment analysis showed that heparin could help in treating sepsis/COVID-19 through immunomodulation, inhibition of the inflammatory response, regulation of angiogenesis and antiviral activity. The pharmacological effects of heparin against these targets were further confirmed by molecular docking and simulation analysis, suggesting that heparin exerts effective binding capacity by targeting the essential residues in sepsis/COVID-19. Prospective clinical practice evaluations may consider the use of these key prognostic indicators for the treatment of sepsis/COVID-19.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yitian Fang
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Peng Cheng Laboratory, Shenzhen, Guangdong, China
| | - Shenggeng Lin
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Qingli Dou
- Department of Emergency Medicine, Affiliated Baoan Hospital of Shenzhen, Southern Medical University, Shenzhen, Guangdong, China
| | - Jianjun Gui
- Department of Emergency Medicine, Affiliated Baoan Hospital of Shenzhen, Southern Medical University, Shenzhen, Guangdong, China
| | - Weimin Li
- National Tuberculosis Clinical Lab of China, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Hongsheng Tan
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanjing Wang
- Engineering Research Center of Cell and Therapeutics Antibody, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Jumei Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Abbas Khan
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Peng Cheng Laboratory, Shenzhen, Guangdong, China
| |
Collapse
|
57
|
Nogueira RS, Salu BR, Nardelli VG, Bonturi CR, Pereira MR, de Abreu Maffei FH, Cilli EM, Oliva MLV. A snake venom-analog peptide that inhibits SARS-CoV-2 and papain-like protease displays antithrombotic activity in mice arterial thrombosis model, without interfering with bleeding time. Thromb J 2023; 21:1. [PMID: 36593467 PMCID: PMC9806807 DOI: 10.1186/s12959-022-00436-5] [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: 07/05/2022] [Accepted: 11/18/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND (p-BthTX-I)2 K, a dimeric analog peptide derived from the C-terminal region of phospholipase A2-like bothropstoxin-I (p-BthTX-I), is resistant to plasma proteolysis and inhibits severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) strains with weak cytotoxic effects. Complications of SARS-CoV-2 infection include vascular problems and increased risk of thrombosis; therefore, studies to identify new drugs for treating SARS-CoV-2 infections that also inhibit thrombosis and minimize the risk of bleeding are required. OBJECTIVES To determine whether (p-BthTX-I)2 K affects the hemostatic system. METHODS Platelet aggregation was induced by collagen, arachidonic acid, and adenosine diphosphate (ADP) in the Chronolog Lumi-aggregometer. The coagulation activity was evaluated by determining activated partial thromboplastin clotting time (aPTT) and prothrombin time (PT) with (p-BthTX-I)2 K (5.0-434.5 µg) or 0.9% NaCl. Arterial thrombosis was induced with a 540 nm laser and 3.5-20 mg kg- 1 Rose Bengal in the carotid artery of male C57BL/6J mice using (p-BthTX-I)2 K. Bleeding time was determined in mouse tails immersed in saline at 37 °C after (p-BthTX-I)2 K (4.0 mg/kg and 8.0 mg/kg) or saline administration. RESULTS (p-BthTX-I)2 K prolonged the aPTT and PT by blocking kallikrein and FXa-like activities. Moreover, (p-BthTX-I)2 K inhibited ADP-, collagen-, and arachidonic acid-induced platelet aggregation in a dose-dependent manner. Further, low concentrations of (p-BthTX-I)2 K extended the time to artery occlusion by the formed thrombus. However, (p-BthTX-I)2 K did not prolong the bleeding time in the mouse model of arterial thrombosis. CONCLUSION These results demonstrate the antithrombotic activity of the peptide (p-BthTX-I)2 K possibly by kallikrein inhibition, suggesting its strong biotechnological potential.
Collapse
Affiliation(s)
- Ruben Siedlarczyk Nogueira
- grid.411249.b0000 0001 0514 7202Department of Biochemistry, Universidade Federal de São Paulo (UNIFESP), SP 04044- 020 São Paulo, Brazil
| | - Bruno Ramos Salu
- grid.411249.b0000 0001 0514 7202Department of Biochemistry, Universidade Federal de São Paulo (UNIFESP), SP 04044- 020 São Paulo, Brazil
| | - Vinícius Goulart Nardelli
- grid.411249.b0000 0001 0514 7202Department of Biochemistry, Universidade Federal de São Paulo (UNIFESP), SP 04044- 020 São Paulo, Brazil
| | - Camila Ramalho Bonturi
- grid.411249.b0000 0001 0514 7202Department of Biochemistry, Universidade Federal de São Paulo (UNIFESP), SP 04044- 020 São Paulo, Brazil
| | - Marina Rodrigues Pereira
- grid.410543.70000 0001 2188 478XDepartment of Biochemistry and Organic Chemistry, Institute of Chemistry, Universidade Estadual Paulista (UNESP), SP 14800-060 São Paulo, Araraquara, Brazil
| | - Francisco Humberto de Abreu Maffei
- grid.410543.70000 0001 2188 478XDepartment of Surgery and Orthopedics, Universidade Estadual Paulista (UNESP), 18618-687 São Paulo, Botucatu, SP Brazil
| | - Eduardo Maffud Cilli
- grid.410543.70000 0001 2188 478XDepartment of Biochemistry and Organic Chemistry, Institute of Chemistry, Universidade Estadual Paulista (UNESP), SP 14800-060 São Paulo, Araraquara, Brazil
| | - Maria Luiza Vilela Oliva
- grid.411249.b0000 0001 0514 7202Department of Biochemistry, Universidade Federal de São Paulo (UNIFESP), SP 04044- 020 São Paulo, Brazil
| |
Collapse
|
58
|
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]
|
59
|
Al-Kuraishy HM, Al-Gareeb AI, Al-Harcan NAH, Alexiou A, Batiha GES. Tranexamic Acid and Plasminogen/Plasmin Glaring Paradox in COVID-19. Endocr Metab Immune Disord Drug Targets 2023; 23:35-45. [PMID: 35927893 DOI: 10.2174/1871530322666220801102402] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/27/2022] [Accepted: 03/30/2022] [Indexed: 11/22/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by a severe acute respiratory syndrome, coronavirus type 2 (SARS-CoV-2), leading to acute tissue injury and an overstated immune response. In COVID-19, there are noteworthy changes in the fibrinolytic system with the development of coagulopathy. Therefore, modulation of the fibrinolytic system may affect the course of COVID-19. Tranexamic acid (TXA) is an anti-fibrinolytic drug that reduces the conversion of plasminogen to plasmin, which is necessary for SARS-CoV-2 infectivity. In addition, TXA has anti-inflammatory, anti-platelet, and anti-thrombotic effects, which may attenuate the COVID-19 severity. Thus, in this narrative review, we try to find the beneficial and harmful effects of TXA in COVID-19.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, AL-Mustansiriyiah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, AL-Mustansiriyiah University, Baghdad, Iraq
| | - Nasser A Hadi Al-Harcan
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Rasheed University College, Baghdad, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia.,AFNP Med Austria, Wien, Austria
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| |
Collapse
|
60
|
Arefizadeh R, Moosavi SH, Towfiqie S, Mohsenizadeh SA, Pishgahi M. Effect of Ticagrelor Compared to Clopidogrel on Short-term Outcomes of COVID-19 Patients with Acute Coronary Syndrome Undergoing Percutaneous Coronary Intervention; a Randomized Clinical Trial. ARCHIVES OF ACADEMIC EMERGENCY MEDICINE 2023; 11:e14. [PMID: 36620733 PMCID: PMC9807941 DOI: 10.22037/aaem.v11i1.1870] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Introduction Acute COVID-19 infection is associated with increased adverse clinical outcomes in patients with acute coronary syndromes (ACS). Given that some studies suggested improved pulmonary function with Ticagrelor, this clinical trial aimed to compare the effects of Ticagrelor versus Clopidogrel on the short-term outcomes of these patients. Methods In this multicenter clinical trial, 180 COVID-19 patients with ACS who underwent urgent percutaneous coronary intervention (PCI) were randomized to receive Ticagrelor (180mg loading dose followed by 90mg twice daily, n=90) or Clopidogrel (600mg loading dose with 75mg daily, n=90), and then followed for one month after their procedure. The primary composite endpoint was a combination of all-cause mortality, myocardial infarction, and early stent thrombosis within the first month after stent implantation. Results After thirty days of follow-up, the primary composite endpoint was non-significantly lower in the Ticagrelor compared to the Clopidogrel group (18.5% vs 23.5% respectively, p = 0.254). Based on the time-to-event analysis, the mean survival rate was 26.8 ±7.7 and 24.7 ±9.9 days, respectively, for the Ticagrelor and the Clopidogrel arms (Log-rank p = 0.275). Secondary endpoints were similar in the two trial arms, except for the mean oxygen saturation, which was higher in the Ticagrelor group (95.28 ±2.68 % vs. 94.15 ± 3.55 %, respectively; p = 0.021). Conclusion Among COVID-19 patients with concomitant ACS, who were treated with urgent PCI, the composite outcome of death, myocardial infarction, and early stent thrombosis was not different between Ticagrelor and Clopidogrel groups. However, administration of Ticagrelor was associated with a slight but statistically significant increase in oxygen saturation compared to Clopidogrel, but this difference wasn't clinically important.
Collapse
Affiliation(s)
- Reza Arefizadeh
- Department of Cardiology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran.,Corresponding author: Reza Arefizadeh; AJA university of medical sciences, Etemad-Zadeh st., Fatemi-Gharbi st., Tehran, Iran. , Tel: +982186096356
| | - Seyed Hossein Moosavi
- Department of Cardiology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Sayied Towfiqie
- Department of Cardiology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | | | - Mehdi Pishgahi
- Department of Cardiology, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Corresponding author: Mehdi Pishgahi; Department of Cardiology, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.Tel:00989123387486, , ORCID: https://orcid.org/0000-0002-1196-6535
| |
Collapse
|
61
|
Smer A, Squires RW, Bonikowske AR, Allison TG, Mainville RN, Williams MA. Cardiac Complications of COVID-19 Infection and the Role of Physical Activity. J Cardiopulm Rehabil Prev 2023; 43:8-14. [PMID: 35839441 PMCID: PMC9828583 DOI: 10.1097/hcr.0000000000000701] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Since December 2019, the newly emerging coronavirus has become a global pandemic with >250 million people infected and >5 million deaths worldwide. Infection with coronavirus disease-2019 (COVID-19) causes a severe immune response and hypercoagulable state leading to tissue injury, organ damage, and thrombotic events. It is well known that COVID-19 infection predominately affects the lungs; however, the cardiovascular complications of the disease have been a major cause of morbidity and mortality. In addition, patients with cardiovascular disease are vulnerable to contract a severe form of the illness and increased mortality. A significant number of patients who survived the disease may experience post-COVID-19 syndrome with a variety of symptoms and physical limitations. Here, we review the cardiac complications of COVID-19 infection and the results of cardiopulmonary exercise testing and guidelines for exercise training after infection.
Collapse
Affiliation(s)
- Aiman Smer
- Department of Cardiovascular Medicine, CHI Health Creighton University School of Medicine, Omaha, Nebraska (Drs Smer and Williams); and Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (Drs Squires, Bonikowske, and Allison and Ms Mainville)
| | - Ray W. Squires
- Department of Cardiovascular Medicine, CHI Health Creighton University School of Medicine, Omaha, Nebraska (Drs Smer and Williams); and Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (Drs Squires, Bonikowske, and Allison and Ms Mainville)
| | - Amanda R. Bonikowske
- Department of Cardiovascular Medicine, CHI Health Creighton University School of Medicine, Omaha, Nebraska (Drs Smer and Williams); and Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (Drs Squires, Bonikowske, and Allison and Ms Mainville)
| | - Thomas G. Allison
- Department of Cardiovascular Medicine, CHI Health Creighton University School of Medicine, Omaha, Nebraska (Drs Smer and Williams); and Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (Drs Squires, Bonikowske, and Allison and Ms Mainville)
| | - Rylie N. Mainville
- Department of Cardiovascular Medicine, CHI Health Creighton University School of Medicine, Omaha, Nebraska (Drs Smer and Williams); and Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (Drs Squires, Bonikowske, and Allison and Ms Mainville)
| | - Mark A. Williams
- Department of Cardiovascular Medicine, CHI Health Creighton University School of Medicine, Omaha, Nebraska (Drs Smer and Williams); and Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (Drs Squires, Bonikowske, and Allison and Ms Mainville)
| |
Collapse
|
62
|
Guest PC, Kesharwani P, Butler AE, Sahebkar A. The COVID-19 Pandemic: SARS-CoV-2 Structure, Infection, Transmission, Symptomology, and Variants of Concern. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1412:3-26. [PMID: 37378759 DOI: 10.1007/978-3-031-28012-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Since it was first detected in December 2019, the COVID-19 pandemic has spread across the world and affected virtually every country and territory. The pathogen driving this pandemic is SARS-CoV-2, a positive-sense single-stranded RNA virus which is primarily transmissible though the air and can cause mild to severe respiratory infections in humans. Within the first year of the pandemic, the situation worsened with the emergence of several SARS-CoV-2 variants. Some of these were observed to be more virulent with varying capacities to escape the existing vaccines and were, therefore, denoted as variants of concern. This chapter provides a general overview of the course of the COVID-19 pandemic up to April 2022 with a focus on the structure, infection, transmission, and symptomology of the SARS-CoV-2 virus. The main objectives were to investigate the effects of the variants of concern on the trajectory of the virus and to highlight a potential pathway for coping with the current and future pandemics.
Collapse
Affiliation(s)
- Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya, Bahrain
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, WA, Australia
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
63
|
Bizjak DA, John L, Matits L, Uhl A, Schulz SVW, Schellenberg J, Peifer J, Bloch W, Weiß M, Grüner B, Bracht H, Steinacker JM, Grau M. SARS-CoV-2 Altered Hemorheological and Hematological Parameters during One-Month Observation Period in Critically Ill COVID-19 Patients. Int J Mol Sci 2022; 23:15332. [PMID: 36499657 PMCID: PMC9735540 DOI: 10.3390/ijms232315332] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/24/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Hematological and hemorheological parameters are known to be altered in COVID-19; however, the value of combined monitoring in order to deduce disease severity is only scarcely examined. A total of 44 acute SARS-CoV-2-infected patients (aCOV) and 44 age-matched healthy controls (Con) were included. Blood of aCOV was sampled at admission (T0), and at day 2 (T2), day 5 (T5), day 10 (T10), and day 30 (T30) while blood of Con was only sampled once. Inter- and intra-group differences were calculated for hematological and hemorheological parameters. Except for mean cellular volume and mean cellular hemoglobin, all blood cell parameters were significantly different between aCOV and Con. During the acute disease state (T0-T5), hematological and hemorheological parameters were highly altered in aCOV; in particular, anemic conditions and increased immune cell response/inflammation, oxidative/nitrosative stress, decreased deformability, as well as increased aggregation, were observed. During treatment and convalescence until T30, almost all abnormal values of aCOV improved towards Con values. During the acute state of the COVID-19 disease, the hematological, as well as the hemorheological system, show fast and potentially pathological changes that might contribute to the progression of the disease, but changes appear to be largely reversible after four weeks. Measuring RBC deformability and aggregation, as well as oxidative stress induction, may be helpful in monitoring critically ill COVID-19 patients.
Collapse
Affiliation(s)
| | - Lucas John
- Division of Sports and Rehabilitation Medicine, University Hospital Ulm, 89075 Ulm, Germany
| | - Lynn Matits
- Division of Sports and Rehabilitation Medicine, University Hospital Ulm, 89075 Ulm, Germany
- Clinical & Biological Psychology, Institute of Psychology and Education, Ulm University, 89081 Ulm, Germany
| | - Alisa Uhl
- Division of Sports and Rehabilitation Medicine, University Hospital Ulm, 89075 Ulm, Germany
| | | | - Jana Schellenberg
- Division of Sports and Rehabilitation Medicine, University Hospital Ulm, 89075 Ulm, Germany
| | - Johannes Peifer
- Central Emergency Services, University Hospital Ulm, 89081 Ulm, Germany
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sports Medicine, Molecular and Cellular Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany
| | - Manfred Weiß
- Clinic for Anaesthesiology and Intensive Care Medicine, University Hospital Medical School, 89081 Ulm, Germany
| | - Beate Grüner
- Department of Internal Medicine III, Division of Infectious Diseases, University Hospital Ulm, 89081 Ulm, Germany
| | - Hendrik Bracht
- Central Emergency Services, University Hospital Ulm, 89081 Ulm, Germany
| | | | - Marijke Grau
- Institute of Cardiovascular Research and Sports Medicine, Molecular and Cellular Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany
| |
Collapse
|
64
|
Vio R, Giordani AS, Stefil M, Alturki A, Russo V, China P, Gasperetti A, Schiavone M, Čulić V, Biondi-Zoccai G, Themistoclakis S, Lip GY, Proietti R. Therapeutic vs. prophylactic anticoagulation in COVID-19 patients: a systematic review and meta-analysis of real-world studies. Minerva Cardiol Angiol 2022; 70:652-662. [PMID: 36305780 DOI: 10.23736/s2724-5683.22.06230-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Coagulopathy, in the form of either venous or arterial thromboembolism, is one of the most severe sequelae of coronavirus disease (COVID-19) and has been associated with poorer outcomes. However, the role of therapeutic anticoagulation (tAC) or prophylactic anticoagulation (pAC) in COVID-19 patients has not been definitely established. Therefore, the aim of this systematic review and meta-analysis was to gather all the available real-world data in the field and to provide a reliable effect size of the effect on mortality of tAC compared to pAC in COVID-19 patients. EVIDENCE ACQUISITION Real-world studies (RWS) were identified by searching electronic databases from inception to 31st October, 2021. Randomized controlled trials were excluded. Mortality and bleedings were considered as primary and secondary outcomes, respectively. EVIDENCE SYNTHESIS 10 RWS and 5541 patients were included in the analysis. Overall, tAC was associated with lower mortality (HR=0.62, 95% CI: 0.54-0.71). There was asymmetry at the funnel plot suggesting publication bias, that was not confirmed at the Egger test (P=0.07). For the secondary endpoint, there was a non-statistically significant tendency for more bleedings in patients treated with tAC compared to pAC (RR=1.75, 95% CI: 0.81-3.81). CONCLUSIONS Our meta-analysis, based on RWS and adjusted estimates of risk, suggests a survival benefit of tAC over pAC in COVID-19 patients in the real world.
Collapse
Affiliation(s)
- Riccardo Vio
- Department of Cardiothoracic, Vascular Medicine and Intensive Care, Dell'Angelo Hospital, Mestre, Venice, Italy -
| | - Andrea S Giordani
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Maria Stefil
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, UK
| | - Ahmed Alturki
- Division of Cardiology, McGill University Health Center, Montreal, Canada
| | - Vincenzo Russo
- Department of Medical Translational Sciences, Monaldi Hospital, Luigi Vanvitelli University of Campania, Naples, Italy
| | - Paolo China
- Department of Cardiothoracic, Vascular Medicine and Intensive Care, Dell'Angelo Hospital, Mestre, Venice, Italy
| | - Alessio Gasperetti
- Unit of Cardiology, ASST Fatebenefratelli Sacco-Luigi Sacco University Hospital, Milan, Italy
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marco Schiavone
- Unit of Cardiology, ASST Fatebenefratelli Sacco-Luigi Sacco University Hospital, Milan, Italy
| | - Viktor Čulić
- School of Medicine, University of Split, Split, Croatia
- Department of Cardiology and Angiology, University Hospital Centre Split, Split, Croatia
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University, Latina, Italy
- Mediterranea Cardiocentro, Naples, Italy
| | - Sakis Themistoclakis
- Department of Cardiothoracic, Vascular Medicine and Intensive Care, Dell'Angelo Hospital, Mestre, Venice, Italy
| | - Gregory Y Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, UK
| | - Riccardo Proietti
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, UK
| |
Collapse
|
65
|
Badr EAE, El Sayed IE, Gabber MKR, Ghobashy EAE, Al-Sehemi AG, Algarni H, Elghobashy YAS. Are Antisense Long Non-Coding RNA Related to COVID-19? Biomedicines 2022; 10:2770. [PMID: 36359290 PMCID: PMC9687826 DOI: 10.3390/biomedicines10112770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/10/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Fighting external pathogens relies on the tight regulation of the gene expression of the immune system. Ferroptosis, which is a distinct form of programmed cell death driven by iron, is involved in the enhancement of follicular helper T cell function during infection. The regulation of RNA is a key step in final gene expression. The present study aimed to identify the expression level of antisense lncRNAs (A2M-AS1, DBH-AS1, FLVCR1-DT, and NCBP2AS2-1) and FLVCR1 in COVID-19 patients and its relation to the severity of the disease. COVID-19 patients as well as age and gender-matched healthy controls were enrolled in this study. The expression level of the antisense lncRNAs was measured by RT-PCR. Results revealed the decreased expression of A2M-AS1 and FLVCR1 in COVID-19 patients. Additionally, they showed the increased expression of DBH-AS1, FLVCR1-DT, and NCBP2AS2. Both FLVCR1-DT and NCBP2AS2 showed a positive correlation with interleukin-6 (IL-6). DBH-AS1 and FLVCR1-DT had a significant association with mortality, complications, and mechanical ventilation. A significant negative correlation was found between A2M-AS1 and NCBP2AS2-1 and between FLVCR1 and FLVCR1-DT. The study confirmed that the expression level of the antisense lncRNAs was deregulated in COVID-19 patients and correlated with the severity of COVID-19, and that it may have possible roles in the pathogenesis of this disease.
Collapse
Affiliation(s)
- Eman A E Badr
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Menoufia University, Shebeen El-Kom 32511, Egypt
| | | | | | | | - Abdullah G. Al-Sehemi
- Research Centre for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
- Department of Chemistry, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Hamed Algarni
- Research Centre for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
- Department of Physics, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Yasser AS Elghobashy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Menoufia University, Shebeen El-Kom 32511, Egypt
| |
Collapse
|
66
|
Mihaljevic O, Zivancevic-Simonovic S, Cupurdija V, Marinkovic M, Tubic Vukajlovic J, Markovic A, Stanojevic-Pirkovic M, Milosevic-Djordjevic O. DNA damage in peripheral blood lymphocytes of severely ill COVID-19 patients in relation to inflammatory markers and parameters of hemostasis. Mutagenesis 2022; 37:203-212. [PMID: 35524945 PMCID: PMC9129204 DOI: 10.1093/mutage/geac011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/11/2022] [Indexed: 01/08/2023] Open
Abstract
Bearing in the mind that a variety of agents can contribute to genome instability, including viral infections, the aim of this study was to analyze DNA damage in hospitalized COVID-19 patients and its relationship with certain laboratory parameters. The potential impact of applied therapy and chest X-rays on DNA damage was also estimated. The study population included 24 severely COVID-19 patients and 15 healthy control subjects. The level of DNA damage was measured as genetic damage index (GDI) by comet assay. The standard laboratory methods and certified enzymatic reagents for the appropriate autoanalyzers were performed for the determination of the biochemical and hematological parameters. COVID-19 patients had significantly higher level of DNA damage compared with control subjects. The absolute number of neutrophil leukocytes was statistically higher, while the absolute number of lymphocytes was statistically lower in COVID-19 patients than in healthy controls. The analysis of the relationship between DNA damage and laboratory parameters indicated that GDI was positively correlated with interleukin 6 (IL-6) concentration and negatively with platelet count in COVID-19 patients. The level of DNA damage was slightly higher in female patients, in whom it was demonstrated a positive correlation of GDI with C-reactive protein (CRP) and procalcitonin. Likewise, there was a negative relationship of GDI and platelet count, and positive relationship of GDI and activated partial thromboplastin time (aPTT) in female population. The applied therapy (antibiotics, corticosteroid, anticoagulant, and antiviral therapy) as well as chest X rays has been shown to have genotoxic potential. The level of DNA damage significantly corresponds to the inflammatory markers and parameters of hemostasis in COVID-19 patients. In conclusion, inflammation, smoking habit, applied therapy, and chest X rays contribute to a higher level of DNA damage in COVID-19 patients.
Collapse
Affiliation(s)
- Olgica Mihaljevic
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | - Vojislav Cupurdija
- Department of Internal Medicine Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Internal medicine, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Milos Marinkovic
- Department of Internal medicine, University Clinical Center Kragujevac, Kragujevac, Serbia
| | | | - Aleksandra Markovic
- Department of Biology, Faculty of Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | - Olivera Milosevic-Djordjevic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Biology, Faculty of Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
67
|
Kaiafa G, Savopoulos C, Karlafti E, Pantazi K, Paramythiotis D, Thomaidou E, Daios S, Ztriva E, Gionis M, Fyntanidou V, Argiriadou H, Didangelos T. Coagulation Profile of COVID-19 Patients. Life (Basel) 2022; 12:1658. [PMID: 36295093 PMCID: PMC9604860 DOI: 10.3390/life12101658] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 12/09/2023] Open
Abstract
Coronavirus disease is a viral infection that can affect multiple systems and be expressed with many-or no-symptoms. The viral infection begins when the virus binds to the host's receptor and from that point on, it is transmitted to the rest of the body, where it causes inflammatory reactions. Among other tissues and systems, SARS-CoV-2 impacts the coagulation system, where it triggers the immunothrombotic response. Its effects are rather intense and can lead to many complications. COVID-19-associated coagulopathy is frequently observed in hospitalized patients, especially ICU patients, and can be proven detrimental. It is usually accompanied by other complications, such as sepsis-induced coagulopathy, disseminated intravascular coagulation and venous thromboembolism. Since all these conditions lead to poor prognosis for severely ill patients, thromboprophylaxis and coagulopathy prognosis are just as important as the therapeutic handling of these patients. Since the beginning of the pandemic, many biomarkers have been considered useful when trying to assess the thrombotic risk of hospitalized patients or evaluate the severity of their situation. At the same time, many drugs have already been tested-while others are still being trialed-in order to find the optimal therapy for each urgent situation.
Collapse
Affiliation(s)
- Georgia Kaiafa
- 1st Propaedeutic Department of Internal Medicine, AHEPA University General Hospital, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Christos Savopoulos
- 1st Propaedeutic Department of Internal Medicine, AHEPA University General Hospital, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Eleni Karlafti
- 1st Propaedeutic Department of Internal Medicine, AHEPA University General Hospital, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
- Emergency Department, AHEPA University General Hospital, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Konstantina Pantazi
- 1st Propaedeutic Department of Internal Medicine, AHEPA University General Hospital, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Daniel Paramythiotis
- 1st Propaedeutic Surgery Department, AHEPA General University Hospital, 54621 Thessaloniki, Greece
| | - Evanthia Thomaidou
- Department of Anesthesia and Intensive Care, AHEPA University General Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Stylianos Daios
- 1st Propaedeutic Department of Internal Medicine, AHEPA University General Hospital, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Eleftheria Ztriva
- 1st Propaedeutic Department of Internal Medicine, AHEPA University General Hospital, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Michalis Gionis
- Vascular Surgery Department, General Hospital of Thessaloniki AHEPA, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Varvara Fyntanidou
- Emergency Department, AHEPA University General Hospital, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Helena Argiriadou
- Department of Anesthesia and Intensive Care, AHEPA University General Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Triantafyllos Didangelos
- 1st Propaedeutic Department of Internal Medicine, AHEPA University General Hospital, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| |
Collapse
|
68
|
Jankauskaite L, Malinauskas M, Snipaitiene A. Effect of stimulated platelets in COVID-19 thrombosis: Role of alpha7 nicotinic acetylcholine receptor. Front Cardiovasc Med 2022; 9:1037369. [PMID: 36312286 PMCID: PMC9614055 DOI: 10.3389/fcvm.2022.1037369] [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: 09/05/2022] [Accepted: 09/26/2022] [Indexed: 01/08/2023] Open
Abstract
Since early 2020, SARS-CoV-2-induced infection resulted in global pandemics with high morbidity, especially in the adult population. COVID-19 is a highly prothrombotic condition associated with subsequent multiorgan failure and lethal outcomes. The exact mechanism of the prothrombotic state is not well understood and might be multifactorial. Nevertheless, platelets are attributed to play a crucial role in COVID-19-associated thrombosis. To date, platelets' role was defined primarily in thrombosis and homeostasis. Currently, more focus has been set on their part in inflammation and immunity. Moreover, their ability to release various soluble factors under activation as well as internalize and degrade specific pathogens has been highly addressed in viral research. This review article will discuss platelet role in COVID-19-associated thrombosis and their role in the cholinergic anti-inflammatory pathway. Multiple studies confirmed that platelets display a hyperactivated phenotype in COVID-19 patients. Critically ill patients demonstrate increased platelet activation markers such as P-selectin, PF4, or serotonin. In addition, platelets contain acetylcholine and express α7 nicotinic acetylcholine receptors (α7nAchR). Thus, acetylcholine can be released under activation, and α7nAchR can be stimulated in an autocrine manner and support platelet function. α7 receptor is one of the most important mediators of the anti-inflammatory properties as it is associated with humoral and intrinsic immunity and was demonstrated to contribute to better outcomes in COVID-19 patients when under stimulation. Hematopoietic α7nAchR deficiency increases platelet activation and, in experimental studies, α7nAchR stimulation can diminish the pro-inflammatory state and modulate platelet reactiveness via increased levels of NO. NO has been described to inhibit platelet adhesion, activation, and aggregation. In addition, acetylcholine has been demonstrated to decrease platelet aggregation possibly by blocking the e p-38 pathway. SARS-CoV-2 proteins have been found to be similar to neurotoxins which can bind to nAChR and prevent the action of acetylcholine. Concluding, the platelet role in COVID-19 thrombotic events could be explained by their active function in the cholinergic anti-inflammatory pathway.
Collapse
Affiliation(s)
- Lina Jankauskaite
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania,Department of Pediatrics, Medical Faculty, Lithuanian University of Health Sciences, Kaunas, Lithuania,*Correspondence: Lina Jankauskaite
| | - Mantas Malinauskas
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ausra Snipaitiene
- Department of Pediatrics, Medical Faculty, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
69
|
Panati K, Timmana LV, Reddy AT V, Reddy Saddala R, Ramireddy Narala V. Virology and Molecular Pathogenesis of Coronavirus Disease 2019: An Update. Eurasian J Med 2022; 54:299-304. [PMID: 35971283 PMCID: PMC9797742 DOI: 10.5152/eurasianjmed.2022.21133] [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: 04/02/2021] [Accepted: 07/06/2021] [Indexed: 01/08/2023] Open
Abstract
The pandemic coronavirus disease 2019 outbreak's causative agent was identified as severe acute respiratory syndrome coronavirus 2. It is a positive-sense single-stranded RNA virus with a ~30 kb size genome that belongs to the Nidovirales. Molecular analysis revealed that severe acute respiratory syndrome coronavirus 2 is a variant of severe acute respiratory syndrome coronavirus and Middle East respiratory syndrome coronavirus with some sequence similarity. The confirmed cases and death toll are high in severe acute respiratory syndrome coronavirus 2 compared to severe acute respiratory syndrome coronavirus and the estimated R0 is >1. The data on pathological findings on severe acute respiratory syndrome coronavirus 2 are scarce and present treatment management is based on symptoms that are similar to severe acute respiratory syndrome coronavirus. In this review, we have discussed the transmission, viral replication, and cytokine storm and highlighted the recent pathological findings of coronavirus disease 2019. The reported severe acute respiratory syndrome coronavirus 2 pathological findings were similar to that of severe acute respiratory syndrome coronavirus. Though these findings help notify the clinical course of the disease, it warrants further in vivo and ex vivo studies with larger samples obtained from the coronavirus disease 2019 patients.
Collapse
Affiliation(s)
- Kalpana Panati
- Department of Biotechnology, Government College for Men, Kadapa, A.P, India
| | - Lokesh V Timmana
- Department of Zoology, Yogi Vemana University, Kadapa, A.P, India
| | | | | | | |
Collapse
|
70
|
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
|
71
|
Kirkan EF, Ulgur HS, Comert S, Erol C, Yildirak MK, Ozkan OF. Management of Spontaneous Major Rectus Sheath Hematoma in a COVID-19 Patient: A Case Report and Literature Review. Cureus 2022; 14:e29206. [PMID: 36258938 PMCID: PMC9569153 DOI: 10.7759/cureus.29206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2022] [Indexed: 12/03/2022] Open
Abstract
A rectus sheath hematoma, which is mostly encountered due to abdominal traumas or anticoagulant use, can be challenging, and a delayed diagnosis may lead to hypovolemic shock and even death. In this study, we aimed to present the management of a case of rectus sheath hematoma that developed in a patient who was hospitalized and under coronavirus disease 2019 (COVID-19) treatment. A 70-year-old female patient was admitted to the intensive care unit (ICU) due to respiratory failure and developed a sudden onset of tachycardia and hypotension. The patient was then diagnosed with a rectus sheath hematoma and after ensuring hemodynamic stability she was treated with angiographic embolization. Following the treatment, the patient remained hemodynamically stable and a control computed tomography (CT) revealed regression in the hematoma. Rectus sheath hematomas especially accompanied by additional comorbidities or aggressive surgical interventions may result in high mortality rates in the early period. It should also be kept in mind that during the COVID-19 pandemic, which has affected the world in the last two years, rectus sheath hematomas may be the underlying cause of sudden hypotension and abdominal distension, and it should not be forgotten that angiographic embolization performed by experienced interventional radiologists is the mainstay of treatment in cases where hemodynamic stability can’t be achieved.
Collapse
|
72
|
Sudusinghe D, Riddell A, Gandhi T, Chowdary P, Davenport A. Increased risk of dialysis circuit clotting in hemodialysis patients with COVID-19 is associated with elevated FVIII, fibrinogen and D-dimers. Hemodial Int 2022; 27:38-44. [PMID: 36081392 PMCID: PMC9537782 DOI: 10.1111/hdi.13046] [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: 03/01/2022] [Revised: 08/09/2022] [Accepted: 08/23/2022] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Severe COVID-19 infections increase the risk of thrombotic events and Intensive Care Units reported increased extracorporeal circuit clotting (ECC) in COVID-19 patients with acute kidney injury. We wished to determine whether hemodialysis (HD) patients with COVID-19 also have increased risk of circuit clotting. METHODS We reviewed coagulation studies and HD records, 4 weeks before and after COVID-19 polymerase chain reaction detection in HD patients between April 2020 and June 2021. FINDINGS Sixty-eight (33.5%) of 203 HD patients with COVID-19, 65% male, mean age 64.9 ± 15.3 years, experienced some circuit clotting, and no clotting recorded prior to positive test results. In those who experienced ECC, prothrombin, activated partial thromboplastin or thrombin times were not different, whereas median factor VIII (273 [168-419] vs. 166 [139-225] IU/dl, p < 0.001), D-dimers (2654 [1381-6019] vs. 1351 [786-2334] ng/ml, p < 0.05), and fibrinogen (5.6 ± 1.4 vs. 4.9 ± 1.4 g/L, p < 0.05) were greater. Antithrombin (94 [83-112] vs. 89 [84-103] IU/dl), protein C (102 [80-130] vs. 86 [76-106] IU/dl), protein S (65 [61-75] vs. 65 [52-79] IU/dl) and platelet counts (193 [138-243] vs. 174 [138-229] × 109 /L) did not differ. On multivariable logistic analysis, circuit clotting was associated with log factor VIII (odds ratio [OR] 14.8 (95% confidence limits [95% CL] 1.12-19.6), p = 0.041), fibrinogen (OR 1.57 [95% CL 1.14-21.7], p = 0.006) and log D dimer (OR 4.8 [95% CL 1.16-12.5], p = 0.028). DISCUSSION Extracorporeal circuit clotting was increased within 4 weeks of testing positive for COVID-19. Clotting was associated with increased factor VIII, fibrinogen and D-dimer, suggesting that the risk of circuit clotting was related to the inflammatory response to COVID-19.
Collapse
Affiliation(s)
- Dinesha Sudusinghe
- Department of Physiology, Faculty of Medical SciencesUniversity of Sri JayewardenepuraNugegodaSri Lanka
| | - Anne Riddell
- Haemophilia and Thrombosis Laboratory (Health Services Laboratories)Royal Free HospitalLondonUK,Katharine Dormandy Haemophilia and Thrombosis CentreRoyal Free HospitalLondonUK
| | - Tejas Gandhi
- Haemophilia and Thrombosis Laboratory (Health Services Laboratories)Royal Free HospitalLondonUK
| | - Pratima Chowdary
- Katharine Dormandy Haemophilia and Thrombosis CentreRoyal Free HospitalLondonUK,Research Department of HaematologyCancer Institute UCLLondonUK
| | - Andrew Davenport
- Department of Renal Medicine, Royal Free Hospital, Faculty of Medical SciencesUniversity College LondonLondonUK
| |
Collapse
|
73
|
Kovács EH, Tánczos K, Szabó L, Turan C, Dembrovszky F, Ocskay K, Lee BY, Hegyi P, Molnár Z. Higher Dose Anticoagulation Cannot Prevent Disease Progression in COVID-19 Patients: A Systematic Review and Meta-Analysis. Biomedicines 2022; 10:2194. [PMID: 36140295 PMCID: PMC9496532 DOI: 10.3390/biomedicines10092194] [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: 08/15/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
Implementation of higher dose (HD) thromboprophylaxis has been considered in patients infected with coronavirus disease 2019 (COVID-19). Our aim was to compare HD to standard dose (SD) thromboprophylaxis in COVID-19 patients. The protocol is registered on PROSPERO (CRD42021284808). We searched for randomised controlled studies (CENTRAL, Embase, Medline and medRxviv) that compared HD to SD anticoagulation in COVID-19 and analysed outcomes such as mortality, thrombotic events, bleedings, and disease progression. The statistical analyses were made using the random effects model. Fourteen articles were included (6253 patients). HD compared with SD showed no difference in mortality (OR 0.83 [95% CI 0.54−1.28]). The use of HD was associated with a decreased risk of thrombosis (OR 0.58 [95% CI 0.44−0.76]), although with an increased risk of major bleeding (OR 1.64 [95% CI 1.25−2.16]). The cohort with D-dimer < 1 mg/mL showed no effect (OR 1.19 [95% CI 0.67−2.11]), but in the case of D-dimer > 1 mg/mL, a tendency of lower risk in the HD group was observed (OR 0.56 [95% CI 0.31−1.00]). The need for intubation in moderately ill patients showed a nonsignificant lower likelihood in the HD group (OR 0.82 [95% CI 0.63−1.08]). We cannot advocate for HD in all COVID-19 patients, although it shows some nonsignificant benefits on disease progression in those with elevated D-dimer who do not need ICU admission.
Collapse
Affiliation(s)
- Emőke Henrietta Kovács
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, 1085 Budapest, Hungary
- Selye János Doctoral College for Advanced Studies, Semmelweis University, 1085 Budapest, Hungary
| | - Krisztián Tánczos
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, 1085 Budapest, Hungary
| | - László Szabó
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Caner Turan
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, 1085 Budapest, Hungary
| | - Fanni Dembrovszky
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Klementina Ocskay
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Bo-Young Lee
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
- Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, 1085 Budapest, Hungary
| | - Zsolt Molnár
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, 1085 Budapest, Hungary
- Department of Anaesthesiology and Intensive Therapy, Faculty of Medicine, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| |
Collapse
|
74
|
Mohamed Y, El-Maradny YA, Saleh AK, Nayl AA, El-Gendi H, El-Fakharany EM. A comprehensive insight into current control of COVID-19: Immunogenicity, vaccination, and treatment. Biomed Pharmacother 2022; 153:113499. [PMID: 36076589 PMCID: PMC9343749 DOI: 10.1016/j.biopha.2022.113499] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 02/07/2023] Open
Abstract
The healthy immune system eliminates pathogens and maintains tissue homeostasis through extraordinarily complex networks with feedback systems while avoiding potentially massive tissue destruction. Many parameters influence humoral and cellular vaccine responses, including intrinsic and extrinsic, environmental, and behavioral, nutritional, perinatal and administrative parameters. The relative contributions of persisting antibodies and immune memory as well as the determinants of immune memory induction, to protect against specific diseases are the main parameters of long-term vaccine efficacy. Natural and vaccine-induced immunity and monoclonal antibody immunotherapeutic, may be evaded by SARS-CoV-2 variants. Besides the complications of the production of COVID-19 vaccinations, there is no effective single treatment against COVID-19. However, administration of a combined treatment at different stages of COVID-19 infection may offer some cure assistance. Combination treatment of antiviral drugs and immunomodulatory drugs may reduce inflammation in critical COVID-19 patients with cytokine release syndrome. Molnupiravir, remdesivir and paxlovid are the approved antiviral agents that may reduce the recovery time. In addition, immunomodulatory drugs such as lactoferrin and monoclonal antibodies are used to control inflammatory responses in their respective auto-immune conditions. Therefore, the widespread occurrence of highly transmissible variants like Delta and Omicron indicates that there is still a lot of work to be done in designing efficient vaccines and medicines for COVID-19. In this review, we briefly discussed the immunological response against SARS-CoV-2 and the vaccines approved by the World Health Organization (WHO) for COVID-19, their mechanisms, and side effects. Moreover, we mentioned various treatment trials and strategies for COVID-19.
Collapse
Affiliation(s)
- Yasser Mohamed
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria 21934, Egypt; Laboratory of Kafr El-Sheikh Fever Hospital, Kafr El-Sheikh Fever Hospital, 33511 Kafr El-Sheikh, Egypt.
| | - Yousra A El-Maradny
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria 21934, Egypt; Microbiology Department, High Institute of Public Health, Alexandria University, Alexandria 21526, Egypt.
| | - Ahmed K Saleh
- Cellulose and Paper Department, National Research Centre, El-Tahrir St., Dokki, P.O. 12622, Giza, Egypt
| | - AbdElAziz A Nayl
- Department of Chemistry, College of Science, Jouf University, Sakaka 72341, Al Jouf, Saudi Arabia.
| | - Hamada El-Gendi
- Bioprocess Development Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), Universities and Research Institutes zone, New Borg El-Arab, Alexandria 21934, Egypt.
| | - Esmail M El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria 21934, Egypt.
| |
Collapse
|
75
|
Apaydin H, Erden A, Güven SC, Armağan B, Konak HE, Polat B, Afşin Y, Kaygisiz M, Omma A, Kucuksahin O. Effects of anti-SARS-CoV-2 vaccination on safety and disease exacerbation in patients with Behçet syndrome in a monocentric cohort. Int J Rheum Dis 2022; 25:1068-1077. [PMID: 35851753 PMCID: PMC9349705 DOI: 10.1111/1756-185x.14387] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/21/2022] [Accepted: 06/29/2022] [Indexed: 01/05/2023]
Abstract
AIM Vaccination represents a cornerstone in mastering the coronavirus disease 2019 (COVID-19) pandemic. There is a paucity of data regarding the safety of COVID-19 vaccines in patients with rheumatic diseases such as Behçet syndrome (BS). The present study aimed to investigate the side-effects and post-vaccine disease exacerbation rates of COVID-19 vaccines in a BS cohort. METHODS We retrospectively evaluated 450 BS patients followed in our clinic who met the criteria of the International Study Group. COVID-19 vaccination status, type of vaccine received (Pfizer-BioNTech vs CoronaVac), post-vaccine side-effects and exacerbations were evaluated by interviewing patients over the phone or face to face. Behçet's Disease Current Activity Form (BDCAF) scores were calculated for BS symptoms before and after vaccination. RESULTS In all, 287 patients received at least one dose of the COVID-19 vaccine. Of the total number of COVID-19 vaccines (n = 639), 379 (59%) were Pfizer-BioNTech vaccines and 257 (41%) were CoronaVac vaccines. The number of side-effects after first, second, third and fourth vaccine doses were 151 (52.6%), 135 (49.4%), 29 (42.6%), and 3 (30%), respectively. BS exacerbation after first, second, third, and fourth vaccine doses were 151 (52.6%), 135 (49.4%), 16 (23.5%), and 3 (30%), respectively. Injection site pain/swelling was the most common side-effect at all vaccine doses followed by fatigue and arthralgia. CONCLUSION COVID-19 vaccines are well tolerated in patients with BS, and more side-effects develop after mRNA vaccines. Regardless of the vaccine type, exacerbations after the COVID-19 vaccine are common, predominantly mucocutaneous and articular involvement, and exacerbations in the form of other organ involvement are rare.
Collapse
Affiliation(s)
- Hakan Apaydin
- Clinical RheumatologyAnkara City HospitalCankayaTurkey
| | - Abdulsamet Erden
- Division of Rheumatology, Department of Internal MedicineYıldırım Beyazıt University, Ankara City HospitalAnkaraTürkiye
| | | | | | | | | | - Yağnur Afşin
- Internal MedicineAnkara City HospitalAnkaraTürkiye
| | | | - Ahmet Omma
- Clinical RheumatologyUniversity of Health SciencesAnkaraTürkiye
| | - Orhan Kucuksahin
- Division of Rheumatology, Department of Internal MedicineYıldırım Beyazıt University, Ankara City HospitalAnkaraTürkiye
| |
Collapse
|
76
|
Rodriguez M, Dai W, Lund H, Osinski K, Zhang Z, Silverstein R, Zheng Z. The correlations among racial/ethnic groups, hypertriglyceridemia, thrombosis, and mortality in hospitalized patients with COVID-19. Best Pract Res Clin Haematol 2022; 35:101386. [PMID: 36494155 PMCID: PMC9527214 DOI: 10.1016/j.beha.2022.101386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 01/08/2023]
Abstract
Reports of racial and ethnic disparities regarding both rates of infection of the SARS-CoV-2 virus and morbidity of the coronavirus disease-19 (COVID-19) contain profound differences depending on the population. Our previous study has shown that patients with COVID-19 who developed hypertriglyceridemia during hospitalization have a 2.3 times higher mortality rate. However, whether the correlation between hypertriglyceridemia and mortality has disparity among different racial and ethnic groups is unknown. In this study, we investigated the impact of race/ethnicity on the correlation between hypertriglyceridemia and mortality in hospitalized patients with COVID-19. De-identified information from 904 hospitalized patients diagnosed with COVID-19 between March 2020 and June 2021 were extracted from the Medical College of Wisconsin Clinical Data Warehouse. A multivariable regression analysis suggested that the Asians and non-White Hispanics had 4 or 3.9 times higher mortality rate, respectively, after adjusting for age, morbid obesity (BMI ≥40), and gender. The hypertriglyceridemia (≥150 mg/dL) was associated with higher mortality, after adjusting for age, gender, and morbid obesity. The baseline hypertriglyceridemia occurrence had relevantly more consistent percentages among all racial/ethnic groups. However, non-White Hispanic and Asian patients had the highest frequencies of peak hypertriglyceridemia occurrence during hospitalization. The peak hypertriglyceridemia developed during hospitalization correlates with the incidence of thrombosis after adjusting for morbid obesity, age, and sex. In summary, in this retrospective study of 904 hospitalized COVID-19 patients, Asians and non-White Hispanics had a greater likelihood of developing hypertriglyceridemia during hospitalization and mortality than White patients.
Collapse
Affiliation(s)
- Maya Rodriguez
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, USA,Diversity Summer Health-Related Research Education Program (DSHREP), Medical College of Wisconsin, Milwaukee, WI, USA,College of Arts and Sciences, Marquette University, Milwaukee, WI, USA
| | - Wen Dai
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, USA,Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hayley Lund
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kristen Osinski
- Clinical & Translational Science Institute (CTSI) Biomedical Informatics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ziyu Zhang
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, USA,Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Roy Silverstein
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, USA,Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ze Zheng
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, USA,Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA,Corresponding author. Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA
| |
Collapse
|
77
|
Comparison of Angiographic and Clinical Outcomes After Primary Percutaneous Coronary Intervention for ST-elevation Myocardial Infarction Between Patients With and Without Concomitant COVID-19 Infection. Crit Pathw Cardiol 2022; 21:141-146. [PMID: 35994723 DOI: 10.1097/hpc.0000000000000297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE COVID-19 infection can involve the cardiovascular system and worsen the prognosis of the patients. This study aimed to investigate the adverse effects of COVID-19 on angiographic and clinical outcomes of primary percutaneous coronary intervention (PCI) in patients with acute ST-elevation MI and compare results with those patients without COVID-19 disease. METHODS The study was a retrospective observational cohort, in which patients presented with ST-elevation MI from February 2020 to April 2021, treated with primary PCI were divided into 2 groups based on the COVID-19 infection. Then, the procedural and angiographic indices and also clinical outcomes were compared between the 2 groups. RESULTS A total of 1150 patients were enrolled in the study. Those with established COVID-19 infection had worse baseline thrombolysis in myocardial infarction flow grade and also were at higher risk for worse procedural outcomes such as lower thrombolysis in myocardial infarction frame count, myocardial blush grade, and slow-flow coronary disease, after the primary PCI. Additionally, the presence of COVID-19 at the time of primary PCI was related to a significantly higher duration of hospitalization and in-hospital mortality. Given the potential impact of other factors on outcomes, analysis for all of the primary endpoints was done again after adjustment of these factors and the results were the same as before, suggesting the independent effect of COVID-19 infection. CONCLUSIONS The concomitant COVID-19 infection in the patients undergoing primary PCI is associated with significantly worse angiographic, procedural and clinical outcomes. Surprisingly, this finding is regardless of patients' baseline risk factors and demographical characteristics.
Collapse
|
78
|
Bonaffini PA, Franco PN, Bonanomi A, Giaccherini C, Valle C, Marra P, Norsa L, Marchetti M, Falanga A, Sironi S. Ischemic and hemorrhagic abdominal complications in COVID-19 patients: experience from the first Italian wave. Eur J Med Res 2022; 27:165. [PMID: 36045452 PMCID: PMC9428880 DOI: 10.1186/s40001-022-00793-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 08/06/2022] [Indexed: 11/10/2022] Open
Abstract
PURPOSE To report ischemic and haemorrhagic abdominal complications in a series of COVID-19 patients. To correlate these complications with lung involvement, laboratory tests, comorbidities, and anticoagulant treatment. METHODS We retrospectively included 30 COVID-19 patients who undergone abdomen CECT for abdominal pain, between March 16 and May 19, 2020. Ischemic and haemorrhagic complications were compared with lung involvement (early, progressive, peak or absorption stage), blood coagulation values, anticoagulant therapy, comorbidities, and presence of pulmonary embolism (PE). RESULTS Ischemic complications were documented in 10 patients (7 receiving anticoagulant therapy, 70%): 6/10 small bowel ischemia (1 concomitant obstruction, 1 perforation) and 4/10 ischemic colitis. Main mesenteric vessels were patent except for 1 superior mesenteric vein thrombosis. Two ischemia cases also presented splenic infarctions. Bleeding complications were found in 20 patients (all receiving anticoagulant treatments), half with active bleeding: hematomas in soft tissues (15) and retroperitoneum (2) and gastro-intestinal bleeding (3). Platelet and lymphocyte were within the normal range. D-Dimer was significantly higher in ischemic cases (p < 0.001). Most of the patients had severe lung disease (45% peak, 29% absorption), two patients PE. CONCLUSIONS Ischemic and haemorrhagic abdominal complications may occur in COVID-19 patients, particularly associated to extended lung disease. CT plays a key role in the diagnosis of these potentially life- threatening conditions.
Collapse
Affiliation(s)
- Pietro Andrea Bonaffini
- Department of Radiology, ASST Papa Giovanni XXIII Hospital, Piazza OMS 1, 24127, Bergamo, BG, Italy. .,School of Medicine, University of Milan-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milan, MI, Italy.
| | - Paolo Niccolò Franco
- Department of Radiology, ASST Papa Giovanni XXIII Hospital, Piazza OMS 1, 24127, Bergamo, BG, Italy.,School of Medicine, University of Milan-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milan, MI, Italy
| | - Alice Bonanomi
- Department of Radiology, ASST Papa Giovanni XXIII Hospital, Piazza OMS 1, 24127, Bergamo, BG, Italy.,School of Medicine, University of Milan-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milan, MI, Italy
| | - Cinzia Giaccherini
- Unit of Immuno-Hematology and Transfusion Medicine and Center of Hemostasis and Thrombosis, Papa Giovanni XXIII Hospital, Piazza OMS 1, 24127, Bergamo, BG, Italy
| | - Clarissa Valle
- Department of Radiology, ASST Papa Giovanni XXIII Hospital, Piazza OMS 1, 24127, Bergamo, BG, Italy.,School of Medicine, University of Milan-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milan, MI, Italy
| | - Paolo Marra
- Department of Radiology, ASST Papa Giovanni XXIII Hospital, Piazza OMS 1, 24127, Bergamo, BG, Italy.,School of Medicine, University of Milan-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milan, MI, Italy
| | - Lorenzo Norsa
- Unit of Pediatric Hepatology Gastroenterology and Transplantation, Papa Giovanni XXIII Hospital, Piazza OMS 1, 24127, Bergamo, BG, Italy
| | - Marina Marchetti
- Unit of Immuno-Hematology and Transfusion Medicine and Center of Hemostasis and Thrombosis, Papa Giovanni XXIII Hospital, Piazza OMS 1, 24127, Bergamo, BG, Italy
| | - Anna Falanga
- School of Medicine, University of Milan-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milan, MI, Italy.,Unit of Immuno-Hematology and Transfusion Medicine and Center of Hemostasis and Thrombosis, Papa Giovanni XXIII Hospital, Piazza OMS 1, 24127, Bergamo, BG, Italy
| | - Sandro Sironi
- Department of Radiology, ASST Papa Giovanni XXIII Hospital, Piazza OMS 1, 24127, Bergamo, BG, Italy.,School of Medicine, University of Milan-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milan, MI, Italy
| |
Collapse
|
79
|
Tudoran C, Velimirovici DE, Berceanu-Vaduva DM, Rada M, Voiţă-Mekeres F, Tudoran M. Increased Susceptibility for Thromboembolic Events versus High Bleeding Risk Associated with COVID-19. Microorganisms 2022; 10:1738. [PMID: 36144340 PMCID: PMC9505654 DOI: 10.3390/microorganisms10091738] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
The infection with the SARS-CoV-2 virus is associated with numerous systemic involvements. Besides the severe respiratory injuries and cardiovascular complications, it became obvious early on that this disease carries an increased risk of thromboembolic events, but a higher propensity for bleedings as well. We researched the medical literature over significant PubMed published articles debating on the prevalence, category of patients, the moment of occurrence, and evolution of venous thromboembolism (VTE), but also of venous and arterial "in situ" thrombosis (AT), and hemorrhagic events as well. Most researchers agree on an increased prevalence of thromboembolic events, ranging between 25 and 31% for VTE, depending on the analyzed population. For AT and hemorrhagic complications lower rates were reported, namely, about 2-3%, respectively, between 4.8 and 8%, occurring mostly in older patients, suffering from moderate/severe forms of COVID-19, with associated comorbidities. It is important to mention that patients suffering from hemorrhages frequently received thromboprophylaxis with anticoagulant drugs. As a consequence of thromboembolic and hemorrhagic complications which are both important negative prognostic factors, the evolution of patients infected with the SARS-CoV-2 virus is aggravated, determining an augmented morbidity and mortality of this population.
Collapse
Affiliation(s)
- Cristina Tudoran
- Department VII, Internal Medicine II, Discipline of Cardiology, University of Medicine and Pharmacy “Victor Babes” Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
- Center of Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, University of Medicine and Pharmacy “Victor Babes” Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
- County Emergency Hospital “Pius Brinzeu”, L. Rebreanu, Nr. 156, 300723 Timisoara, Romania
- Academy of Romanian Scientists, Ilfov Str. Nr. 3, 50085 Bucuresti, Romania
| | - Dana Emilia Velimirovici
- Department VI, Internal Medicine and Ambulatory Care, Prevention and Cardiovascular Recovery, University of Medicine and Pharmacy “Victor Babes” Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
| | - Delia Mira Berceanu-Vaduva
- Department XIV, Microbiology, University of Medicine and Pharmacy “Victor Babes” Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
| | - Maria Rada
- Department VI, Internal Medicine and Ambulatory Care, Prevention and Cardiovascular Recovery, University of Medicine and Pharmacy “Victor Babes” Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
| | - Florica Voiţă-Mekeres
- Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 1 Universitatii Street, 410087 Oradea, Romania
| | - Mariana Tudoran
- County Emergency Hospital “Pius Brinzeu”, L. Rebreanu, Nr. 156, 300723 Timisoara, Romania
| |
Collapse
|
80
|
Alrashed A, Cahusac P, Mohzari YA, Bamogaddam RF, Alfaifi M, Mathew M, Alrumayyan BF, Alqahtani BF, Alshammari A, AlNekhilan K, Binrokan A, Alamri K, Alshahrani A, Alshahrani S, Alanazi AS, Alhassan BM, Alsaeed A, Almutairi W, Albujaidy A, AlJuaid L, Almalki ZS, Ahmed N, Alajami HN, Aljishi HM, Alsheef M, Alajlan SA, Almutairi F, Alsirhani A, Alotaibi M, Aljaber MA, Bahammam HA, Aldandan H, Almulhim AS, Abraham I, Alamer A. A comparison of three thromboprophylaxis regimens in critically ill COVID-19 patients: An analysis of real-world data. Front Cardiovasc Med 2022; 9:978420. [PMID: 36051287 PMCID: PMC9424612 DOI: 10.3389/fcvm.2022.978420] [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: 06/26/2022] [Accepted: 07/18/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Thrombotic complications of coronavirus disease 2019 (COVID-19) have received considerable attention. Although numerous conflicting findings have compared escalated thromboprophylaxis doses with a standard dose to prevent thrombosis, there is a paucity of literature comparing clinical outcomes in three different anticoagulation dosing regimens. Thus, we investigated the effectiveness and safety profiles of standard, intermediate, and high-anti-coagulation dosing strategies in COVID-19 critically ill patients. Methodology This retrospective multicenter cohort study of intensive care unit (ICU) patients from the period of April 2020 to August 2021 in four Saudi Arabian centers. Inclusion criteria were age ≥ 18 years, diagnosis with severe or critical COVID-19 infection, and receiving prophylactic anticoagulant dose within 24-48 h of ICU admission. The primary endpoint was a composite of thrombotic events, with mortality rate and minor or major bleeding serving as secondary endpoints. We applied survival analyses with a matching weights procedure to control for confounding variables in the three arms. Results A total of 811 patient records were reviewed, with 551 (standard-dose = 192, intermediate-dose = 180, and high-dose = 179) included in the analysis. After using weights matching, we found that the standard-dose group was not associated with an increase in the composite thrombotic events endpoint when compared to the intermediate-dose group {19.8 vs. 25%; adjusted hazard ratio (aHR) =1.46, [95% confidence of interval (CI), 0.94-2.26]} or when compared to high-dose group [19.8 vs. 24%; aHR = 1.22 (95% CI, 0.88-1.72)]. Also, there were no statistically significant differences in overall in-hospital mortality between the standard-dose and the intermediate-dose group [51 vs. 53.4%; aHR = 1.4 (95% CI, 0.88-2.33)] or standard-dose and high-dose group [51 vs. 61.1%; aHR = 1.3 (95% CI, 0.83-2.20)]. Moreover, the risk of major bleeding was comparable in all three groups [standard vs. intermediate: 4.8 vs. 2.8%; aHR = 0.8 (95% CI, 0.23-2.74); standard vs. high: 4.8 vs. 9%; aHR = 2.1 (95% CI, 0.79-5.80)]. However, intermediate-dose and high-dose were both associated with an increase in minor bleeding incidence with aHR = 2.9 (95% CI, 1.26-6.80) and aHR = 3.9 (95% CI, 1.73-8.76), respectively. Conclusion Among COVID-19 patients admitted to the ICU, the three dosing regimens did not significantly affect the composite of thrombotic events and mortality. Compared with the standard-dose regimen, intermediate and high-dosing thromboprophylaxis were associated with a higher risk of minor but not major bleeding. Thus, these data recommend a standard dose as the preferred regimen.
Collapse
Affiliation(s)
- Ahmed Alrashed
- Administration of Pharmaceutical Services, Main Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Peter Cahusac
- Pharmacology and Biostatistics/Comparative Medicine, Alfaisal University College of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Yahya A. Mohzari
- Clinical Pharmacy Department, King Saud Medical City, Riyadh, Saudi Arabia
| | - Reem F. Bamogaddam
- Clinical Pharmacy Department, King Saud Medical City, Riyadh, Saudi Arabia
| | - Mashael Alfaifi
- Clinical Pharmacy Department, King Saud Medical City, Riyadh, Saudi Arabia
| | - Maya Mathew
- Clinical Pharmacy Department, King Saud Medical City, Riyadh, Saudi Arabia
| | - Bashayer F. Alrumayyan
- Administration of Pharmaceutical Services, Main Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Basmah F. Alqahtani
- Administration of Pharmaceutical Services, Main Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Amjad Alshammari
- Administration of Pharmaceutical Services, Main Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Kholud AlNekhilan
- Administration of Pharmaceutical Services, Main Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Aljawharah Binrokan
- Administration of Pharmaceutical Services, Main Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Khalil Alamri
- Administration of Pharmaceutical Services, Main Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Abdullah Alshahrani
- Administration of Pharmaceutical Services, Main Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Safar Alshahrani
- Administration of Pharmaceutical Services, Main Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ahmad S. Alanazi
- Administration of Pharmaceutical Services, Main Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Batool M. Alhassan
- Department of Clinical Pharmacy, Almoosa Specialist Hospital, Al-Ahasa, Saudi Arabia
| | - Ali Alsaeed
- Department of Neurology, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | - Asma Albujaidy
- Department of Clinical Pharmacy Service, Prince Mohammed Bin Abdulaziz Hospital, Riyadh, Saudi Arabia
| | - Lama AlJuaid
- Pharmacy College, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ziyad S. Almalki
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Nehad Ahmed
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Hamdan N. Alajami
- Clinical Pharmacy Department, King Saud Medical City, Riyadh, Saudi Arabia
| | - Hala M. Aljishi
- Research Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Mohammed Alsheef
- Medicine Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Saleh A. Alajlan
- Department of Pediatric Dentistry, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Faisal Almutairi
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Atheer Alsirhani
- Department of Pharmacy Service, Prince Mutib Bin Abdulaziz Hospital, Sakaka, Saudi Arabia
| | - Manayer Alotaibi
- Department of Pharmacy Service, Prince Mutib Bin Abdulaziz Hospital, Sakaka, Saudi Arabia
| | - Melaf A. Aljaber
- Department of Pharmacy Service, Prince Mutib Bin Abdulaziz Hospital, Sakaka, Saudi Arabia
| | - Hammam A. Bahammam
- Department of Pediatric Dentistry, College of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Abdulaziz S. Almulhim
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Ivo Abraham
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tuscon, AZ, United States
| | - Ahmad Alamer
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tuscon, AZ, United States
| |
Collapse
|
81
|
Lucignani G, Guarnera A, Rossi-Espagnet MC, Moltoni G, Antonelli A, Figà Talamanca L, Carducci C, Calo Carducci FI, Napolitano A, Gandolfo C, Campi F, Auriti C, Parazzini C, Longo D. From Fetal to Neonatal Neuroimaging in TORCH Infections: A Pictorial Review. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1210. [PMID: 36010101 PMCID: PMC9406729 DOI: 10.3390/children9081210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022]
Abstract
Congenital infections represent a challenging and varied clinical scenario in which the brain is frequently involved. Therefore, fetal and neonatal neuro-imaging plays a pivotal role in reaching an accurate diagnosis and in predicting the clinical outcome. Congenital brain infections are characterized by various clinical manifestations, ranging from nearly asymptomatic diseases to syndromic disorders, often associated with severe neurological symptoms. Brain damage results from the complex interaction among the infectious agent, its specific cellular tropism, and the stage of development of the central nervous system at the time of the maternal infection. Therefore, neuroradiological findings vary widely and are the result of complex events. An early detection is essential to establishing a proper diagnosis and prognosis, and to guarantee an optimal and prompt therapeutic perinatal management. Recently, emerging infective agents (i.e., Zika virus and SARS-CoV2) have been related to possible pre- and perinatal brain damage, thus expanding the spectrum of congenital brain infections. The purpose of this pictorial review is to provide an overview of the current knowledge on fetal and neonatal brain neuroimaging patterns in congenital brain infections used in clinical practice.
Collapse
Affiliation(s)
- Giulia Lucignani
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| | - Alessia Guarnera
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| | | | - Giulia Moltoni
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, 00146 Rome, Italy
| | - Amanda Antonelli
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
- Department of Radiology, IRCCS Materno Infantile Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy
| | - Lorenzo Figà Talamanca
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| | - Chiara Carducci
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| | | | - Antonio Napolitano
- Medical Physics Unit, Risk Management Enterprise, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| | - Carlo Gandolfo
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| | - Francesca Campi
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| | - Cinzia Auriti
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| | - Cecilia Parazzini
- Department of Pediatric Radiology and Neuroradiology, V. Buzzi Children’s Hospital, Via Castelvetro 32, 20154 Milan, Italy
| | - Daniela Longo
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| |
Collapse
|
82
|
The prevalence of vascular complications in SARS-CoV-2 infected outpatients. Wien Med Wochenschr 2022; 173:168-172. [PMID: 35939218 PMCID: PMC9358629 DOI: 10.1007/s10354-022-00954-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022]
Abstract
Background The risk of thromboembolic events is increased for coronavirus disease (COVID)-19 inpatients. For severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected outpatients, only few data are available so far. Methods In our prospective single-center study, 461 SARS-CoV-2-infected outpatients were screened for the presence of deep vein thrombosis. Results Two outpatients had suffered a deep vein thrombosis. An association with previously known risk factors, such as preexisting thrombosis in the medical history or cardiovascular risk factors, could not be proven. Conclusion General thromboprophylaxis in SARS-CoV-2-infected outpatients is still not recommended.
Collapse
|
83
|
Pilia E, Belletti A, Fresilli S, Finco G, Landoni G. Efficacy and safety of heparin full-dose anticoagulation in hospitalized non-critically ill COVID-19 patients: a meta-analysis of multicenter randomized controlled trials. J Thromb Thrombolysis 2022; 54:420-430. [PMID: 35922578 PMCID: PMC9362611 DOI: 10.1007/s11239-022-02681-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2022] [Indexed: 12/24/2022]
Abstract
Arterial and venous thrombotic events in COVID-19 cause significant morbidity and mortality among patients. Although international guidelines agree on the need for anticoagulation, it is unclear whether full-dose heparin anticoagulation confers additional benefits over prophylactic-dose anticoagulation. This systematic review and meta-analysis aimed to investigate the efficacy and safety of heparin full-dose anticoagulation in hospitalized non-critically ill COVID-19 patients. We searched Pubmed/Medline, EMBASE, Clinicaltrials.gov, medRxiv.org and Cochrane Central Register of clinical trials dated up to April 2022. Randomized controlled trials (RCTs) comparing full-dose heparin anticoagulation to prophylactic-dose anticoagulation or standard treatment in hospitalized non-critically ill COVID-19 patients were included in our pooled analysis. The primary endpoint was the rate of major thrombotic events and the co-primary endpoint was the rate of major bleeding events. We identified 4 studies, all of them multicenter, randomizing 2926 patients. Major thrombotic events were 23/1524 (1.5%) in full-dose heparin anticoagulation versus 57/1402 (4.0%) in prophylactic-dose [relative risk (RR) 0.39; 95% confidence interval (CI) 0.25–0.62; p˂0.01; I2 = 0%]. Clinical relevant bleeding events occurred in 1.7% (26/1524) among patients treated with heparin full anticoagulation dose compared to 1.1% (15/1403) in prophylactic-dose group (RR 1.60; 95% CI 0.85–3.03; p = 0.15; I2 = 20%). Mortality was 6.6% (101/1524) versus 8.6% (121/1402) (RR 0.63; 95% CI 0.33–1.19; p = 0.15). In this meta-analysis of high quality multicenter randomized trials, full-dose anticoagulation with heparin was associated with lower rate of major thrombotic events without differences in bleeding risk and mortality in hospitalized non critically ill COVID-19 patients. Study registration PROSPERO, review no. CRD42022301874.
Collapse
Affiliation(s)
- Eros Pilia
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy.,Department of Anesthesia, Resuscitation and Pain Therapy, University of Cagliari, Cagliari, Italy
| | - Alessandro Belletti
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Stefano Fresilli
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Gabriele Finco
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy.,Department of Anesthesia, Resuscitation and Pain Therapy, University of Cagliari, Cagliari, Italy
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy. .,School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
84
|
LaVasseur C, Neukam S, Kartika T, Samuelson Bannow B, Shatzel J, DeLoughery TG. Hormonal therapies and venous thrombosis: Considerations for prevention and management. Res Pract Thromb Haemost 2022; 6:e12763. [PMID: 36032216 PMCID: PMC9399360 DOI: 10.1002/rth2.12763] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 04/12/2022] [Accepted: 05/21/2022] [Indexed: 11/18/2022] Open
Abstract
Background Venous thromboses are well-established complications of hormonal therapy. Thrombosis risk is seen with both hormonal contraceptive agents and with hormone replacement therapy for menopause and gender transition. Over the past several decades, large epidemiological studies have helped better define these risks. Objectives To review and discuss the differences in thrombosis risk of the many of hormonal preparations available as well as their interaction with patient-specific factors. Methods We conducted a narrative review of the available literature regarding venous thrombosis and hormonal therapies including for contraception, menopausal symptoms, and gender transition. Results Thrombosis risk with estrogen-containing compounds increases with increasing systemic dose of estrogen. While progesterone-only-containing products are not associated with thrombosis, when paired with estrogen in combined oral contraceptives, the formulation of progesterone does impact the risk. These components, along with patient-specific factors, may influence the choice of hormonal preparation. For patients who develop thrombosis on hormonal treatment, anticoagulation is protective against future thrombosis. Duration of anticoagulation is dependent on ongoing and future hormone therapy choice. Finally, the optimal management of hormone therapy for individuals diagnosed with prothrombotic illnesses such as COVID-19 remains unclear. Conclusions When contemplating hormonal contraception or hormone replacement therapy, clinicians must consider a variety of factors including hormone type, dose, route, personal and family history of thrombosis, and other prothrombotic risk factors to make informed, personalized decisions regarding the risk of venous thrombosis.
Collapse
Affiliation(s)
- Corinne LaVasseur
- Department of MedicineOregon Health and Sciences UniversityPortlandOregonUSA
| | - Suvi Neukam
- Department of MedicineOregon Health and Sciences UniversityPortlandOregonUSA
- Division of Internal MedicineOregon Health and Sciences UniversityPortlandOregonUSA
| | - Thomas Kartika
- Division of Hematology‐OncologyOregon Health and Sciences UniversityPortlandOregonUSA
| | - Bethany Samuelson Bannow
- Department of MedicineOregon Health and Sciences UniversityPortlandOregonUSA
- Division of Hematology‐OncologyOregon Health and Sciences UniversityPortlandOregonUSA
- The Hemophilia CenterOregon Health and Sciences UniversityPortlandOregonUSA
| | - Joseph Shatzel
- Department of MedicineOregon Health and Sciences UniversityPortlandOregonUSA
- Division of Hematology‐OncologyOregon Health and Sciences UniversityPortlandOregonUSA
| | - Thomas G. DeLoughery
- Department of MedicineOregon Health and Sciences UniversityPortlandOregonUSA
- Division of Hematology‐OncologyOregon Health and Sciences UniversityPortlandOregonUSA
- Division of Laboratory Medicine, Department of PathologyOregon Health and Sciences UniversityPortlandOregonUSA
| |
Collapse
|
85
|
Thomas MR, Scully M. Clinical features of thrombosis and bleeding in COVID-19. Blood 2022; 140:184-195. [PMID: 35452509 PMCID: PMC9040438 DOI: 10.1182/blood.2021012247] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 04/13/2022] [Indexed: 01/08/2023] Open
Abstract
Infection with the SARS-CoV-2 virus, resulting in COVID-19 disease, has presented a unique scenario associated with high rates of thrombosis. The risk of venous thrombosis is some three- to sixfold higher than for patients admitted to a hospital for other indications, and for patients who have thrombosis, mortality appears to increase. Thrombosis may be a presenting feature of COVID-19. Pulmonary thrombi are the most frequent events, some related to deep vein thrombosis, but also to in situ microvascular and macrovascular thrombosis. Other venous thromboses include catheter- and circuit-associated in patients requiring hemofiltration and extracorporeal membrane oxygenation. Arterial thrombosis is less commonly documented, with 3% of patients in intensive care units having major arterial strokes and up to 9% having myocardial infarction, both of which are most likely multifactorial. Risk factors for thrombosis above those already documented in hospital settings include duration of COVID-19 symptoms before admission to the hospital. Laboratory parameters associated with higher risk of thrombosis include higher D-dimer, low fibrinogen, and low lymphocyte count, with higher factor VIII and von Willebrand factor levels indicative of more severe COVID-19 infection. All patients should receive thromboprophylaxis when admitted with COVID-19 infection, but the dose and length of treatment are still debated. Thrombosis continues to be treated according to standard VTE guidelines, but adjustments may be needed depending on other factors relevant to the patient's admission.
Collapse
Affiliation(s)
- Mari R Thomas
- Department of Haematology, University College London Hospital (UCLH), London, United Kingdom; and
- Cardiometabolic Programme, National Institute for Health and Care Research (NIHR), UCLH, University College London Biomedical Research Centre (BRC), London, United Kingdom
| | - Marie Scully
- Department of Haematology, University College London Hospital (UCLH), London, United Kingdom; and
- Cardiometabolic Programme, National Institute for Health and Care Research (NIHR), UCLH, University College London Biomedical Research Centre (BRC), London, United Kingdom
| |
Collapse
|
86
|
Carmona-Rivera C, Zhang Y, Dobbs K, Markowitz TE, Dalgard CL, Oler AJ, Claybaugh DR, Draper D, Truong M, Delmonte OM, Licciardi F, Ramenghi U, Crescenzio N, Imberti L, Sottini A, Quaresima V, Fiorini C, Discepolo V, Lo Vecchio A, Guarino A, Pierri L, Catzola A, Biondi A, Bonfanti P, Poli Harlowe MC, Espinosa Y, Astudillo C, Rey-Jurado E, Vial C, de la Cruz J, Gonzalez R, Pinera C, Mays JW, Ng A, Platt A, Drolet B, Moon J, Cowen EW, Kenney H, Weber SE, Castagnoli R, Magliocco M, Stack MA, Montealegre G, Barron K, Fink DL, Kuhns DB, Hewitt SM, Arkin LM, Chertow DS, Su HC, Notarangelo LD, Kaplan MJ. Multicenter analysis of neutrophil extracellular trap dysregulation in adult and pediatric COVID-19. JCI Insight 2022; 7:160332. [PMID: 35852866 PMCID: PMC9534551 DOI: 10.1172/jci.insight.160332] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/14/2022] [Indexed: 12/02/2022] Open
Abstract
Dysregulation in neutrophil extracellular trap (NET) formation and degradation may play a role in the pathogenesis and severity of COVID-19; however, its role in the pediatric manifestations of this disease, including multisystem inflammatory syndrome in children (MIS-C) and chilblain-like lesions (CLLs), otherwise known as “COVID toes,” remains unclear. Studying multinational cohorts, we found that, in CLLs, NETs were significantly increased in serum and skin. There was geographic variability in the prevalence of increased NETs in MIS-C, in association with disease severity. MIS-C and CLL serum samples displayed decreased NET degradation ability, in association with C1q and G-actin or anti-NET antibodies, respectively, but not with genetic variants of DNases. In adult COVID-19, persistent elevations in NETs after disease diagnosis were detected but did not occur in asymptomatic infection. COVID-19–affected adults displayed significant prevalence of impaired NET degradation, in association with anti-DNase1L3, G-actin, and specific disease manifestations, but not with genetic variants of DNases. NETs were detected in many organs of adult patients who died from COVID-19 complications. Infection with the Omicron variant was associated with decreased NET levels when compared with other SARS-CoV-2 strains. These data support a role for NETs in the pathogenesis and severity of COVID-19 in pediatric and adult patients.
Collapse
Affiliation(s)
- Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
| | - Yu Zhang
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID); and
| | | | | | - Clifton L. Dalgard
- Department of Anatomy, Physiology & Genetics, School of Medicine, and the American Genome Center, Uniformed Services University of the Health Sciences (USUHS), Bethesda, Maryland, USA
| | - Andrew J. Oler
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, Maryland, USA
| | - Dillon R. Claybaugh
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
| | | | | | | | | | - Ugo Ramenghi
- Department of Public Health and Pediatric Sciences and
| | - Nicoletta Crescenzio
- Pediatric Hematology, “Regina Margherita” Children Hospital, University of Turin, Turin, Italy
| | - Luisa Imberti
- Centro di Ricerca Emato-oncologica AIL, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Alessandra Sottini
- Centro di Ricerca Emato-oncologica AIL, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Virginia Quaresima
- Centro di Ricerca Emato-oncologica AIL, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Chiara Fiorini
- Centro di Ricerca Emato-oncologica AIL, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Valentina Discepolo
- Department of Translational Medical Sciences, Pediatric Section, University of Naples Federico II, Naples, Italy
| | - Andrea Lo Vecchio
- Department of Translational Medical Sciences, Pediatric Section, University of Naples Federico II, Naples, Italy
| | - Alfredo Guarino
- Department of Translational Medical Sciences, Pediatric Section, University of Naples Federico II, Naples, Italy
| | - Luca Pierri
- Department of Translational Medical Sciences, Pediatric Section, University of Naples Federico II, Naples, Italy
| | - Andrea Catzola
- Department of Translational Medical Sciences, Pediatric Section, University of Naples Federico II, Naples, Italy
| | - Andrea Biondi
- Department of Pediatrics, University of Milano-Bicocca, European Reference Network (ERN) PaedCan, EuroBloodNet, MetabERN, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Paolo Bonfanti
- Department of Infectious Diseases, San Gerardo Hospital–University of Milano-Bicocca, Monza, Italy
| | - Maria C. Poli Harlowe
- Programa de Inmunogenética e Inmunología Traslacional, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
- Hospital Roberto del Rio, Santiago, Chile
| | | | | | - Emma Rey-Jurado
- Programa de Inmunogenética e Inmunología Traslacional, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Cecilia Vial
- Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Programa Hantavirus, Instituto de Ciencias e Innovación en Medicina, Santiago, Chile
| | - Javiera de la Cruz
- Programa de Inmunogenética e Inmunología Traslacional, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Ricardo Gonzalez
- Pediatric Intensive Care Unit, Hospital Exequiel Gonzalez Cortés, Santiago, Chile
| | - Cecilia Pinera
- Infectious Diseases Unit, Hospital Dr. Exequiel González Cortés, Región Metropolitana, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Jacqueline W. Mays
- National Institute of Dental and Craniofacial Research (NIDCR), NIH, Bethesda, Maryland, USA
| | - Ashley Ng
- Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Andrew Platt
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, and Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA
| | | | | | - Beth Drolet
- Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - John Moon
- Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | | | | | | | | | - Mary Magliocco
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, NIAID; and
| | - Michael A. Stack
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, NIAID; and
| | - Gina Montealegre
- Division of Clinical Research, NIAID, NIH, Bethesda, Maryland, USA
| | - Karyl Barron
- Division of Clinical Research, NIAID, NIH, Bethesda, Maryland, USA
| | - Danielle L. Fink
- Applied/Developmental Research Directorate, Frederick and National Laboratory for Cancer Research, National Cancer Institute (NCI), NIH, Frederick, Maryland, USA
| | - Douglas B. Kuhns
- Applied/Developmental Research Directorate, Frederick and National Laboratory for Cancer Research, National Cancer Institute (NCI), NIH, Frederick, Maryland, USA
| | - Stephen M. Hewitt
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, USA
| | - Lisa M. Arkin
- Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Daniel S. Chertow
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, and Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA
| | - Helen C. Su
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID); and
| | | | - Mariana J. Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
| |
Collapse
|
87
|
Chmiela T, Rzepka M, Kuca M, Serwońska K, Laskowski M, Koperczak A, Siuda J. Intravenous Thrombolysis for Acute Ischemic Stroke during the COVID-19 Pandemic-Polish Single-Center Retrospective Cohort Study. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071068. [PMID: 35888156 PMCID: PMC9321477 DOI: 10.3390/life12071068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022]
Abstract
COVID-19 has affected the entire world and has had a great impact on healthcare, influencing the treatment of patients with acute ischemic stroke (AIS). The aim of this study was to determine the impact of the COVID-19 pandemic on the care of patients with AIS. We performed a retrospective analysis of 1599 patients diagnosed with AIS and hospitalized in the authors' institution from January 2018 to December 2021. The final sample consisted of 265 patients treated with thrombolysis without a diagnosis of COVID-19. The initiation of thrombolytic treatment during the pandemic was delayed (2:42 ± 0:51 vs. 2:25 ± 0:53; p = 0.0006). The delay was mainly related to the pre-hospital phase (1:41 ± 0:48 vs. 1:26 ± 0:49; p = 0.0014), and the door-to-needle time was not affected. There were no differences in stroke severity and patients' outcomes. Patients with AIS were less likely to have previously been diagnosed with atrial fibrillation (16.9% vs. 26.7%; p = 0.0383), ischemic heart disease (25.3% vs. 46.5%; p = 0.0003) and hyperlipidemia (31.2% vs. 46.5%; p = 0.0264). Patients treated during the pandemic had higher glycemia (149.45 ± 54. vs. 143.25 ± 60.71 mg/dL; p= 0.0012), while no significant differences in their lipid profiles were found. Conclusions: The COVID-19 pandemic affected the treatment of AIS patients locally at our stroke center. It caused treatment delay and hindered the recognition of risk factors prior to the occurrence of AIS.
Collapse
Affiliation(s)
- Tomasz Chmiela
- Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (M.R.); (J.S.)
- Correspondence:
| | - Michalina Rzepka
- Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (M.R.); (J.S.)
| | - Maciej Kuca
- Students’ Scientific Association, Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (M.K.); (K.S.); (M.L.); (A.K.)
| | - Karolina Serwońska
- Students’ Scientific Association, Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (M.K.); (K.S.); (M.L.); (A.K.)
| | - Maciej Laskowski
- Students’ Scientific Association, Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (M.K.); (K.S.); (M.L.); (A.K.)
| | - Agnieszka Koperczak
- Students’ Scientific Association, Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (M.K.); (K.S.); (M.L.); (A.K.)
| | - Joanna Siuda
- Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (M.R.); (J.S.)
| |
Collapse
|
88
|
Thromboprophylaxis and clinical outcomes in moderate COVID-19 patients: A comparative study. Res Social Adm Pharm 2022; 18:4048-4055. [PMID: 35864037 PMCID: PMC9288247 DOI: 10.1016/j.sapharm.2022.07.004] [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: 01/22/2022] [Revised: 03/28/2022] [Accepted: 07/10/2022] [Indexed: 01/08/2023]
Abstract
Background Many thrombotic complications are linked to coronavirus disease 2019 (COVID-19). Antithrombotic treatments are important for prophylaxis against these thrombotic events. Objectives This study was designed to compare enoxaparin and rivaroxaban as prophylactic anticoagulants in moderate cases of COVID-19 in terms of efficacy, safety, and clinical outcomes. Methods The study involved 124 patients with moderate COVID-19 (pneumonia without hypoxia) divided into two groups. The first group (G1) comprised 66 patients who received enoxaparin subcutaneously at a dose of 0.5 mg/kg every 12 h until discharge from the hospital. The second group (G2) comprised 58 patients who received oral rivaroxaban at a dose of 10 mg once daily until discharge from the hospital. The outcomes evaluated in this study were as follows: intermediate care unit (IMCU) duration, the number of patients transferred from the IMCU to the intensive care unit (ICU), ICU duration, the total length of hospital stay, in-hospital mortality, and thrombotic and bleeding complications. Results No significant differences in IMCU duration (p = 0.39), ICU duration (p = 0.96), and total length of hospital stay (p = 0.73) were observed between the two groups. The percentage of patients requiring ICU admission after hospitalization was 21.2% in G1 and 22.4% in G2 (p = 0.87). The mortality rate was 12.1% in G1 and 10.3% in G2 (p = 0.76). The proportion of patients who had thrombotic complications was 9.1% in G1 and 12.1% in G2 (p = 0.59). The incidence of mild bleeding was 3% in G1 and 1.7% in G2 (p = 0.64). Conclusion Either enoxaparin or rivaroxaban may be used as thromboprophylaxis agents in managing patients with moderate COVID-19. Either medication has no clear advantage over the other.
Collapse
|
89
|
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
|
90
|
McGill JR, Lagassé HAD, Hernandez N, Hopkins L, Jankowski W, McCormick Q, Simhadri V, Golding B, Sauna ZE. A structural homology approach to identify potential cross-reactive antibody responses following SARS-CoV-2 infection. Sci Rep 2022; 12:11388. [PMID: 35794133 PMCID: PMC9259575 DOI: 10.1038/s41598-022-15225-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/21/2022] [Indexed: 11/09/2022] Open
Abstract
The emergence of the novel SARS-CoV-2 virus is the most important public-health issue of our time. Understanding the diverse clinical presentations of the ensuing disease, COVID-19, remains a critical unmet need. Here we present a comprehensive listing of the diverse clinical indications associated with COVID-19. We explore the theory that anti-SARS-CoV-2 antibodies could cross-react with endogenous human proteins driving some of the pathologies associated with COVID-19. We describe a novel computational approach to estimate structural homology between SARS-CoV-2 proteins and human proteins. Antibodies are more likely to interrogate 3D-structural epitopes than continuous linear epitopes. This computational workflow identified 346 human proteins containing a domain with high structural homology to a SARS-CoV-2 Wuhan strain protein. Of these, 102 proteins exhibit functions that could contribute to COVID-19 clinical pathologies. We present a testable hypothesis to delineate unexplained clinical observations vis-à-vis COVID-19 and a tool to evaluate the safety-risk profile of potential COVID-19 therapies.
Collapse
Affiliation(s)
- Joseph R McGill
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - H A Daniel Lagassé
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Nancy Hernandez
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Louis Hopkins
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Wojciech Jankowski
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Quinn McCormick
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Vijaya Simhadri
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Basil Golding
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Zuben E Sauna
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
91
|
Calabretta F, Preti PS, Russo M, Klersy C, Di Sabatino A. The role of heparin in reducing in-hospital complications and three-month mortality rates in hospitalized COVID-19 patients. Eur J Intern Med 2022; 101:124-127. [PMID: 35307245 PMCID: PMC8923898 DOI: 10.1016/j.ejim.2022.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022]
Affiliation(s)
- F Calabretta
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy.
| | - P S Preti
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - M Russo
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - C Klersy
- Clinical Epidemiology and Biometry Service. IRCCS Policlinico San Matteo Foundation. Pavia, Italy
| | - A Di Sabatino
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| |
Collapse
|
92
|
Duca ŞT, Costache AD, Miftode RŞ, Mitu O, PetriŞ AO, Costache II. Hypercoagulability in COVID-19: from an unknown beginning to future therapies. Med Pharm Rep 2022; 95:236-242. [PMID: 36060499 PMCID: PMC9387574 DOI: 10.15386/mpr-2195] [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: 05/05/2021] [Revised: 05/17/2022] [Accepted: 05/31/2022] [Indexed: 11/23/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a global public health concern and is characterized by an exaggerated inflammatory response that can lead to a large variety of clinical manifestations such as respiratory distress, sepsis, coagulopathy, and death. While it was initially considered primarily a respiratory illness, different data suggests that COVID-19 can lead to a pro-inflammatory milieu and a hypercoagulable state. Several mechanisms attempt to explain the pro-coagulant state seen in COVID-19 patients, including increased fibrinogen concentration, different receptor binding, exhausted fibrinolysis, cytokine storm, and endothelial dysfunction. Some hematological parameters, such as elevated D-dimers and other fibrinolytic products, indicate that the essence of coagulopathy is massive fibrin formation. Moreover, elevated D-dimer levels have emerged as an independent risk factor for a worse outcome, including death, indicating a potential risk for deep vein thrombosis and pulmonary thromboembolism. Prophylactic anticoagulation is recommended in all in-patients with COVID-19 to reduce the incidence of thrombosis. Those with elevated D-dimer values or with a higher risk of developing thromboembolic events should be treated with higher doses of anticoagulant. Anticoagulation may not be enough in some circumstances, highlighting the need for alternative therapies. An understanding of the complex cross-talk between inflammation and coagulopathy is necessary for developing direct appropriate interventional strategies.
Collapse
Affiliation(s)
- Ştefania-Teodora Duca
- Department of Medical Specialties I, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, Romania
- "Sf. Spiridon" County Clinical Emergency Hospital, Iasi, Romania
| | - Alexandru-Dan Costache
- Department of Medical Specialties I, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, Romania
- "Sf. Spiridon" County Clinical Emergency Hospital, Iasi, Romania
| | - Radu-Ştefan Miftode
- Department of Medical Specialties I, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, Romania
- "Sf. Spiridon" County Clinical Emergency Hospital, Iasi, Romania
| | - Ovidiu Mitu
- Department of Medical Specialties I, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, Romania
- "Sf. Spiridon" County Clinical Emergency Hospital, Iasi, Romania
| | - Antoniu-Octavian PetriŞ
- Department of Medical Specialties I, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, Romania
- "Sf. Spiridon" County Clinical Emergency Hospital, Iasi, Romania
| | - Irina-Iuliana Costache
- Department of Medical Specialties I, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, Romania
- "Sf. Spiridon" County Clinical Emergency Hospital, Iasi, Romania
| |
Collapse
|
93
|
Stefanile A, Cellerino M, Koudriavtseva T. Elevated risk of thrombotic manifestations of SARS-CoV-2 infection in cancer patients: A literature review. EXCLI JOURNAL 2022; 21:906-920. [PMID: 36172074 PMCID: PMC9489888 DOI: 10.17179/excli2022-5073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/20/2022] [Indexed: 11/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19) results in higher risks of hospitalization or death in older patients and those with multiple comorbidities, including malignancies. Patients with cancer have greater risks of COVID-19 onset and worse prognosis. This excess is mainly explained by thrombotic complications. Indeed, an imbalance in the equilibrium between clot formation and bleeding, increased activation of coagulation, and endothelial dysfunction characterize both COVID-19 patients and those with cancer. With this review, we provide a summary of the pathological mechanisms of coagulation and thrombotic manifestations in these patients and discuss the possible therapeutic implications of these phenomena.
Collapse
Affiliation(s)
- Annunziata Stefanile
- Department Clinical Pathology and Cancer Biobank, IRCCS Regina Elena National Cancer Institute, Istituti Fisioterapici Ospitalieri (IFO), 00144 Rome, Italy
| | - Maria Cellerino
- Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, IFO, Via Elio Chianesi 53, 00144, Rome, Italy,Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Genoa, Italy,*To whom correspondence should be addressed: Maria Cellerino, Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, IFO, Via Elio Chianesi 53, 00144, Rome, Italy and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Genoa, Italy, E-mail:
| | - Tatiana Koudriavtseva
- Medical Direction, IRCCS Regina Elena National Cancer Institute, Istituti Fisioterapici Ospitalieri (IFO), 00144 Rome, Italy
| |
Collapse
|
94
|
Aissaoui O, Husam S, Mounir A, Benouna EG, Benmallem O, El Kettani C, Barrou L. Concomitant acute limb ischemia and myocardial infarction: another challenge of COVID-19's hypercoagulability. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2022; 12:149-152. [PMID: 35873181 PMCID: PMC9301031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 05/26/2022] [Indexed: 06/15/2023]
Abstract
The COVID-19, actual pandemic due to SARS COV 2 is associated with numerous thromboembolic complications. Although venous thrombosis including pulmonary embolisms have been widely described, arterial localization seems rarely reported. Acute limb ischemia and myocardial infarction are two major consequences of arterial thrombosis and their concomitant occurrence among COVID-19 patients is extremely rare. It is an evident aspect of hypercoagulability and a real challenge to physicians. We herein describe the management of a 77 years old COVID-19 patient presenting an acute lower limb ischemia with concomitant myocardial infarction. He underwent coronary angiography with subsequent stent placement then was transferred to the operating room where a thrombectomy was performed. The outcome was poor as the cardiogenic shock persisted in addition to a reperfusion syndrome with multiorgan failure.
Collapse
Affiliation(s)
- Ouissal Aissaoui
- Department of Intensive Care and Anesthesiology, Ibn Rochd University Hospital, Laboratory of Clinical Immunology, Inflammation and Allergy (LICIA), Hassan II University of CasablancaMorocco
| | - Salem Husam
- Department of Intensive Care and Anesthesiology, Ibn Rochd University Hospital, Laboratory of Clinical Immunology, Inflammation and Allergy (LICIA), Hassan II University of CasablancaMorocco
| | - Anass Mounir
- Department of Intensive Care and Anesthesiology, Ibn Rochd University Hospital, Laboratory of Clinical Immunology, Inflammation and Allergy (LICIA), Hassan II University of CasablancaMorocco
| | - El Ghali Benouna
- Department of Cardiology, Ibn Rochd University Hospital, Hassan II University of CasablancaMorocco
| | - Othmane Benmallem
- Department of Cardiology, Ibn Rochd University Hospital, Hassan II University of CasablancaMorocco
| | - Chafik El Kettani
- Department of Intensive Care and Anesthesiology, Ibn Rochd University Hospital, Laboratory of Clinical Immunology, Inflammation and Allergy (LICIA), Hassan II University of CasablancaMorocco
| | - Lahoucine Barrou
- Department of Intensive Care and Anesthesiology, Ibn Rochd University Hospital, Laboratory of Clinical Immunology, Inflammation and Allergy (LICIA), Hassan II University of CasablancaMorocco
| |
Collapse
|
95
|
Marchioni C, Esposito G, Calci M, Bais B, Colussi G. Effect of intermediate/high versus low dose heparin on the thromboembolic and hemorrhagic risk of unvaccinated COVID-19 patients in the emergency department. BMC Emerg Med 2022; 22:107. [PMID: 35698054 PMCID: PMC9192337 DOI: 10.1186/s12873-022-00668-8] [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: 04/08/2022] [Accepted: 06/10/2022] [Indexed: 11/10/2022] Open
Abstract
Background The optimal prophylactic dose of heparin in patients with coronavirus-associated disease 2019 (COVID-19) in the emergency department (ED) is debated. This study aimed to analyze different thromboprophylaxis approaches in unvaccinated COVID-19 patients admitted to ED without initial venous thromboembolism. Methods Retrospectively, the effect of intermediate/high versus low dose heparin treatment was evaluated from December 2020 to July 2021 in a tertiary Academic Hospital in northeast Italy. The primary outcome comprised arterial or venous thromboembolism or all-cause death within 30 days. Secondary outcomes comprised each single primary outcome component or major hemorrhagic event. Cox regression was used to determine predictors of the primary outcome and propensity score weights to balance the effect of heparin treatment on all outcomes. Results Data of 144 consecutive patients (age 70 ± 13, 33% females) were included in the study. High-dose prophylactic heparin was used in 69%, intermediate in 15%, and low in 17% of patients. The primary outcome occurred in 48 patients. Independent predictors of the primary outcome were COVID-19 severity (hazards ratio (HR) 1.96, 95% confidence interval (CI) 1.05–3.65, p = 0.035) and D-dimer levels (HR each log ng/dl 1.38, 95% CI 1.04–1.84, p = 0.026). Intermediate/high dose heparin did not affect the risk of the primary outcome compared with the low dose (weighted HR 1.39, 95% CI 0.75–2.56, p = 0.292). Intermediate/high heparin increased the risk of major hemorrhagic events (weighted HR 5.92, 95% CI 1.09–32, p = 0.039). Conclusions In unvaccinated COVID-19 patients admitted to ED, prophylaxis with heparin at the intermediate/high dose did not reduce primary outcome compared with the low dose but increased the risk of major hemorrhagic events.
Collapse
Affiliation(s)
- Claudia Marchioni
- Division of Internal Medicine and Emergency Medicine Residency Program, Department of Medicine, University of Udine, 33100, Udine, Italy
| | - Gaetano Esposito
- Department of Emergency Medicine, ASUFC University Hospital of Udine, 33100, Udine, Italy
| | - Mario Calci
- Department of Emergency Medicine, ASUFC University Hospital of Udine, 33100, Udine, Italy
| | - Bruno Bais
- Thrombosis Prevention Unit, 2nd Division of Internal Medicine, Department of Medicine, ASUFC University Hospital of Udine, 33100, Udine, Italy
| | - GianLuca Colussi
- Division of Internal Medicine and Emergency Medicine Residency Program, Department of Medicine, University of Udine, 33100, Udine, Italy.
| |
Collapse
|
96
|
Tjahjadi M, Caropeboka S, Permana C, Susanto K, Susanto E. Long-Delayed Manifestation of COVID-19 Coagulopathy Presenting with Severe Cerebral Venous Thrombosis Causes Massive Brain Hemorrhage. Asian J Neurosurg 2022; 17:342-346. [PMID: 36120629 PMCID: PMC9473853 DOI: 10.1055/s-0042-1750388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
COVID-19 infection causes coagulopathy, which may lead to cerebral venous thrombotic (CVT) event. It usually occurs in patients with higher severity level of infection and manifests mostly within a month after the infection. However, in rare cases, the CVT may happen long after the infection and unrelated to the degree of the infection severity. We present the case of a previously healthy 62-year-old male patient with very mild COVID-19 symptoms that resolved in 3 weeks of home isolation treatment. Immediately after the infection, he developed hypercoagulability and was treated routinely with a novel oral anti-coagulant drug. Four months after the infection, he developed a worsening headache which, in several days, deteriorated to cause reduction in his consciousness level. Imaging showed a right temporoparietooccipital massive brain hemorrhage with right transverse and sigmoid sinus thrombosis. Emergency decompressive craniectomy was performed and the patient recovery was excellent. In patients with a hypercoagulable state after COVID-19 infection, the possibility of CVT event should be observed. It may not be related to the severity of the infection, and it may happen long after the infection.
Collapse
Affiliation(s)
- Mardjono Tjahjadi
- Department of Surgery, Faculty of Medicine, Universitas Katolik Indonesia Atma Jaya, Jakarta, Indonesia
- Department of Neurosurgery, Mitra Keluarga Kemayoran Hospital, Jakarta, Indonesia
| | - Sinatrya Caropeboka
- Department of Neurosurgery, Mitra Keluarga Kemayoran Hospital, Jakarta, Indonesia
| | - Christian Permana
- Department of Neurosurgery, Mitra Keluarga Kemayoran Hospital, Jakarta, Indonesia
| | - Krisandi Susanto
- Department of Neurosurgery, Mitra Keluarga Kemayoran Hospital, Jakarta, Indonesia
| | - Eka Susanto
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
97
|
Alakuş Ü, Kara U, Tasçı C, Eryılmaz M. Upper gastrointestinal system bleedings in COVID-19 patients: Risk factors and management/a retrospective cohort study. ULUS TRAVMA ACIL CER 2022; 28:762-768. [PMID: 35652869 PMCID: PMC10443021 DOI: 10.14744/tjtes.2021.30513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/20/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Upper gastrointestinal system bleeding (UGIB) that occurs with the effect of coagulopathy due to COVID-19 disease itself and drugs such as LMWH and steroids used in the treatment negatively affects the outcomes. In this study, we aimed to examine the frequency of gastrointestinal system bleeding in COVID-19 patients, risk factors, effect on outcomes, and management. METHODS Institutional center (a third-level pandemic center) database was searched for patients hospitalized for COVID-19 between March 11, 2020, and December 17, 2020, retrospectively. Patients with UGIB symptoms/signs were included in the study. Age, gender, body mass index (kg/m2), hospital department where bleeding was diagnosed, previous bleeding history, comorbidities, and medication were steroid, anticoagulant, low weight molecule heparin, and proton-pomp inhibitor, endoscopic findings/treatment, transfusion, and mortality rates were evaluated. Patients were divided into two groups as survivors and non-survivors and parameters were compared. RESULTS Forty-five of a total 5484 patients under COVID-19 treatment had upper gastrointestinal bleeding (0.8%). The average age of the patients was 70.1 years and 73% bleeders were male. Nineteen patients (44%) underwent endoscopy. The most common etiologies of bleeding were gastric/duodenal ulcer (n=9), erosive gastritis (n=4), and hemorrhagic gastritis (n=3). Active bleeding re-quiring intervention was detected in only one patient; therapeutic band ligation was applied to only 1 (2%) of all patients. The most common etiologies of bleeding were gastric/duodenal ulcer (n=9), erosive gastritis (n=4), and hemorrhagic gastritis (n=3). In terms of statistical significance, it was observed that the rate of steroid treatment (77% vs. 39%) and the number of days of steroid treatment were higher in non-survivor group. CONCLUSION UGIB is less common in COVID-19 patients compared to other hospitalized patients. However, it significantly increases mortality. Mortality risk increases even more in patients using steroids. These risks should be considered in patients under COVID-19 treatment. The majority of the bleeding patients does not require endoscopic treatment and should be managed conser-vatively. It is worth considering reducing unnecessary endoscopies in the pandemic.
Collapse
Affiliation(s)
- Ümit Alakuş
- Department of General Surgery, Division of Gastroenterologic Surgery, University of Health Sciences, Gülhane Training and Research Hospital, Ankara-Turkey
| | - Umut Kara
- Department of Anesthesiology and Reanimation, University of Health Sciences, Gülhane Training and Research Hospital, Ankara-Turkey
| | - Cantürk Tasçı
- Department of Pulmonology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara-Turkey
| | - Mehmet Eryılmaz
- Department of General Surgery, University of Health Sciences, Gülhane Training and Research Hospital, Ankara-Turkey
| |
Collapse
|
98
|
Maldonado G, Guerrero R, Intriago M, Rios C. d-Dimer trend in a long-term rheumatoid arthritis patient with tofacitinib treatment and recent COVID-19 infection. REVISTA COLOMBIANA DE REUMATOLOGÍA 2022. [PMCID: PMC8255109 DOI: 10.1016/j.rcreu.2021.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The SARS-CoV-2 virus was first identified in December 2019, the infection was named COVID-19. The initial symptoms and evolution of the disease have been described over the past year. The virus has been shown to increase the risk of thromboembolic events due to the hypercoagulable state triggered by systemic endothelial inflammation. We present the case of a patient with a history of rheumatoid arthritis under prolonged treatment with tofacitinib, who presented COVID-19 and subsequently developed a hypercoagulable state of approximately 6 months’ duration. The possible association between viral infection and the use of tofacitinib is debated.
Collapse
|
99
|
Rajendran R, Chathambath A, Al-Sehemi AG, Pannipara M, Unnikrishnan MK, Aleya L, Raghavan RP, Mathew B. Critical role of nitric oxide in impeding COVID-19 transmission and prevention: a promising possibility. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:38657-38672. [PMID: 35258738 PMCID: PMC8902850 DOI: 10.1007/s11356-022-19148-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 02/06/2022] [Indexed: 05/06/2023]
Abstract
COVID-19 is a serious respiratory infection caused by a beta-coronavirus that is closely linked to SARS. Hypoxemia is a symptom of infection, which is accompanied by acute respiratory distress syndrome (ARDS). Augmenting supplementary oxygen may not always improve oxygen saturation; reversing hypoxemia in COVID-19 necessitates sophisticated means to promote oxygen transfer from alveoli to blood. Inhaled nitric oxide (iNO) has been shown to inhibit the multiplication of the respiratory coronavirus, a property that distinguishes it from other vasodilators. These findings imply that NO may have a crucial role in the therapy of COVID-19, indicating research into optimal methods to restore pulmonary physiology. According to clinical and experimental data, NO is a selective vasodilator proven to restore oxygenation by helping to normalize shunts and ventilation/perfusion mismatches. This study examines the role of NO in COVID-19 in terms of its specific physiological and biochemical properties, as well as the possibility of using inhaled NO as a standard therapy. We have also discussed how NO could be used to prevent and cure COVID-19, in addition to the limitations of NO.
Collapse
Affiliation(s)
- Rajalakshmi Rajendran
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Anjana Chathambath
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Abdullah G Al-Sehemi
- Research Center for Advanced Materials Science, King Khalid University, Abha, 61413, Saudi Arabia
- Department of Chemistry, King Khalid University, Abha, 61413, Saudi Arabia
| | - Mehboobali Pannipara
- Research Center for Advanced Materials Science, King Khalid University, Abha, 61413, Saudi Arabia
- Department of Chemistry, King Khalid University, Abha, 61413, Saudi Arabia
| | | | - Lotfi Aleya
- Laboratoire Chrono-Environment, Universite de Bourgogne Franche-Comte, CNRS6249, Besancon, France
| | - Roshni Pushpa Raghavan
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India.
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, 682 041, India
| |
Collapse
|
100
|
Al-Mamoori HS, Ahmed MH, Al-Nafie TYS, Al-Attar Z. Assessment of the Level of Protein C in Hospitalized Iraqi Patients with COVID-19 and its Correlation with Hematological and Inflammatory Markers. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND: COVID-19 coagulopathy manifests by elevation of certain marker of active coagulation as fibrinogen and this increment associated with increased markers of inflammations.
AIM: To measure protein C (PC) level in hospitalized patients with COVID-19 and to find a possible correlation with hematological and inflammatory markers.
PATIENTS AND METHODS: Seventy-five hospitalized Iraqi adult patients with COVID-19 were included in a descriptive cross-sectional research. PC, D-dimer, and erythrocyte sedimentation rate (ESR) blood samples were collected, and further information was received from patient’s records. Statistical analysis was conducted using SPSS version 23 and Microsoft Office Excel 2019.
RESULTS: Mean age of 75 patients included in the study was 60.13 ± 14.65 years. Sixty-two (62.7%) of patients exhibited neutrophilia, whereas 41 had lymphopenia (54.7%). High ratio of neutrophil/lymphocyte (N/L) was seen in 66 (88.0%), eosinopenia was seen in 46 (61.3%), high lactate dehydrogenase level was seen 68 (90.7%), serum ferritin was high in 66 (88.0%), and high level of C-reactive protein was seen in 68 (90.7%), increased ESR was seen in 69 (92.0%) and high level of D-dimer was seen in 56 (74.7%), while low level of PC was seen in 12 (16.0%) patients. PC had significant negative correlation with prothrombin and partial thromboplastin time but no significant correlation with hematological and inflammatory parameters.
CONCLUSION: COVID-19 coagulopathy is common in majority of patients which include significant changes in WBCs counts, inflammatory markers, PC, and D-dimer levels. Such changes may have a great impact on morbidity and mortality and thus need to be monitored throughout treatment and convalescence.
Collapse
|