1
|
Sabit H, Arneth B, Altrawy A, Ghazy A, Abdelazeem RM, Adel A, Abdel-Ghany S, Alqosaibi AI, Deloukas P, Taghiyev ZT. Genetic and Epigenetic Intersections in COVID-19-Associated Cardiovascular Disease: Emerging Insights and Future Directions. Biomedicines 2025; 13:485. [PMID: 40002898 PMCID: PMC11852909 DOI: 10.3390/biomedicines13020485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/23/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
The intersection of COVID-19 and cardiovascular disease (CVD) has emerged as a significant area of research, particularly in understanding the impact of antiplatelet therapies like ticagrelor and clopidogrel. COVID-19 has been associated with acute cardiovascular complications, including myocardial infarction, thrombosis, and heart failure, exacerbated by the virus's ability to trigger widespread inflammation and endothelial dysfunction. MicroRNAs (miRNAs) play a critical role in regulating these processes by modulating the gene expressions involved in platelet function, inflammation, and vascular homeostasis. This study explores the potential of miRNAs such as miR-223 and miR-126 as biomarkers for predicting resistance or responsiveness to antiplatelet therapies in COVID-19 patients with cardiovascular disease. Identifying miRNA signatures linked to drug efficacy could optimize treatment strategies for patients at high risk of thrombotic events during COVID-19 infection. Moreover, understanding miRNA-mediated pathways offers new insights into how SARS-CoV-2 exacerbates CVD, particularly through mechanisms like cytokine storms and endothelial damage. The findings of this research could lead to personalized therapeutic approaches, improving patient outcomes and reducing mortality in COVID-19-associated cardiovascular events. With global implications, this study addresses the urgent need for effective management of CVD in the context of COVID-19, focusing on the integration of molecular biomarkers to enhance the precision of antiplatelet therapy.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Afaf Altrawy
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Aysha Ghazy
- Department of Agri-Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Rawan M. Abdelazeem
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Amro Adel
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Amany I. Alqosaibi
- Department of Biology, College of Science, Imam Abdulrahman bin Faisal University, Dammam 31441, Saudi Arabia
| | - Panos Deloukas
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK;
| | - Zulfugar T. Taghiyev
- Department of Cardiovascular Surgery, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
2
|
Ozer V, Gonenc Cekic O, Bulbul O, Aydın D, Bulut E, Aksoy F, Pehlivanlar Kucuk M, Caner Karahan S, Emel Sozen E, Ozkaya E, Kosucu P, Karaca Y, Turedi S. Diagnostic and Prognostic Value of SCUBE-1 in COVID-19 Patients. West J Emerg Med 2024; 25:975-984. [PMID: 39625772 PMCID: PMC11610722 DOI: 10.5811/westjem.18586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 12/06/2024] Open
Abstract
Introduction The workload of physicians increased due to the number of patients presenting with suspicion of coronavirus 2019 (COVID-19) and the prolonged wait times in the emergency department during the COVID-19 pandemic. Signal peptide-CUB-EGF domain-containing protein 1 (SCUBE-1) is a protein present in platelets and endothelial cells; it is activated by inflammation from COVID-19 and may be associated with COVID-19's known thrombotic risk. We aimed to determine whether SCUBE-1 levels are diagnostically correlated in suspected COVID-19 patients, and whether SCUBE-1 correlated with severity of disease and, therefore, might be useful to guide hospitalization/discharge decisions. Methods The suspected COVID-19 patients cared for at tertiary healthcare institutions for one year between May 2021-May 2022 were examined in this study. The subjects were both suspected COVID-19 patients not ultimately found to have COVID-19 and those who were diagnosed with COVID-19. By modifying the disease severity scoring systems present in COVID-19 guidelines in 2021, the COVID-19-positive patient group was classified as mild, moderate, severe, and critical, and compared using the SCUBE-1 levels. Moreover, SCUBE-1 levels were compared between the COVID-19 positive group and the COVID-19 negative group. Results A total of 507 patients were considered for the present study. After excluding 175 patients for incomplete data and alternate comorbid organ failure. we report on 332 patients (65.5%). Of these 332 patients, 80 (24.0%) were COVID-19 negative, and 252 (76.0%) were COVID-19 positive. Of 252 (100%) patients diagnosed with COVID-19, 74 (29.4%) were classified as mild, 95 (37.7%) moderate, 45 (17.8%) severe, and 38 (15.1%) critical. The SCUBE-1 levels were statistically different between COVID-19 positive (8.48 ± 7.42 nanograms per milliliter [ng/mL]) and COVID-19 negative (1.86 ± 0.92 ng/mL) patients (P < 0.001). In the COVID-19 positive group, SCUBE-1 levels increased with disease severity (mild = 3.20 ± 1.65 ng/mL, moderate = 4.78 ± 2.26 ng/mL, severe = 13.68 ± 3.95 ng/mL, and critical = 21.87 ± 5.39 ng/mL) (P < 0.001). The initial SCUBE-1 levels of discharged patients were significantly lower than those requiring hospitalization (discharged = 2.89 ng/mL [0.55-8.60 ng/mL]; ward admitted = 7.13 ng/mL [1.38-21.29 ng/mL], and ICU admitted = 21.19 ng/mL [10.58-37.86 ng/mL]) (P < 0.001). Conclusion The SCUBE-1 levels were found to be differentiated between patients with and without COVID-19 and to be correlated with the severity of illness.
Collapse
Affiliation(s)
- Vildan Ozer
- Karadeniz Technical University, School of Medicine, Department of Emergency Medicine, Trabzon, Türkiye
| | - Ozgen Gonenc Cekic
- SBU Kanuni Training and Research Hospital, Department of Emergency Medicine, Trabzon, Türkiye
| | - Ozlem Bulbul
- Karadeniz Technical University, School of Medicine, Department of Emergency Medicine, Trabzon, Türkiye
| | - Davut Aydın
- SBU Kanuni Training and Research Hospital, Department of Chest Diseases, Division of Intensive Care Medicine, Trabzon, Türkiye
| | - Eser Bulut
- SBU Kanuni Training and Research Hospital, Department of Radiology, Trabzon, Türkiye
| | - Firdevs Aksoy
- Karadeniz Technical University, School of Medicine, Department of Infectious Diseases and Clinical Microbiology, Trabzon, Türkiye
| | - Mehtap Pehlivanlar Kucuk
- Karadeniz Technical University, School of Medicine, Department of Chest Diseases, Division of Intensive Care Medicine, Trabzon, Türkiye
| | - Suleyman Caner Karahan
- Karadeniz Technical University, School of Medicine, Department of Biochemistry, Trabzon, Türkiye
| | - Ebru Emel Sozen
- SBU Kanuni Training and Research Hospital, Department of Infectious Diseases and Clinical Microbiology, Trabzon, Türkiye
| | - Esra Ozkaya
- Karadeniz Technical University, School of Medicine, Department of Medical Microbiology, Trabzon, Türkiye
| | - Polat Kosucu
- Karadeniz Technical University, School of Medicine, Department of Radiology, Trabzon, Türkiye
| | - Yunus Karaca
- Karadeniz Technical University, School of Medicine, Department of Emergency Medicine, Trabzon, Türkiye
| | - Suleyman Turedi
- SBU Kanuni Training and Research Hospital, Department of Emergency Medicine, Trabzon, Türkiye
| |
Collapse
|
3
|
Mitsiakos G, Gialamprinou D, Kontovazainitis CG, Moraitis A, Katsaras G, Pouliakis A, Diamanti E. Coagulation assessment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infected pregnant women and their offspring by using rotational thromboelastometry (ROTEM). J Perinat Med 2024; 52:327-342. [PMID: 38353249 DOI: 10.1515/jpm-2023-0444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 03/07/2024]
Abstract
OBJECTIVES During pregnancy, severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection may intensify the gestational procoagulant state. Coronavirus disease 2019 (COVID-19) associated coagulopathy (CAC) constitutes an exacerbated immunothrombosis response. There is limited data regarding the coagulation profile of SARS-CoV2-infected pregnant women, especially those with CAC, and the effect on their offspring. This prospective study aimed to compare the hemostatic profile of those women and their neonates with healthy mother-neonate pairs. METHODS Conventional coagulation tests (CCTs) and rotational thromboelastometry (ROTEM) were employed to evaluate the hemostatic profiles. Neonates were assessed at birth and on the fourth day of life. RESULTS We enrolled 46 SARS-CoV2-infected pregnant women and 22 healthy controls who gave birth to 47 and 22 neonates, respectively. CAC was present in 10 participants. SARS-CoV2-infected pregnant women manifested slightly prolonged APTT and higher fibrinogen levels. Regarding ROTEM, we noted decreased FIBTEM CFT, with higher A10, A-angle, and MCF. The CAC group presented lower platelet count, increased fibrinogen levels, and higher FIBTEM A10 and MCF. PT was slightly prolonged at birth in neonates born to SARS-CoV2-infected mothers. During the fourth day of life, D-dimers were significantly increased. Concerning ROTEM, neonates born to SARS-CoV2-infected mothers showed lower FIBTEM CT at birth. CONCLUSIONS SARS-CoV2-infected pregnant women present a hypercoagulable profile. Hypercoagulability with elevated fibrinolysis and lower platelet count is observed in participants with CAC. The coagulation profile of neonates born to SARS-CoV2 mothers seems unaffected. Elevated D-dimers on the fourth day may reflect a neonatal inflammatory response to maternal SARS-CoV2.
Collapse
Affiliation(s)
- Georgios Mitsiakos
- 2nd Neonatal Department and Neonatal Intensive Care Unit (NICU), Aristotle University of Thessaloniki, "Papageorgiou" Hospital, Thessaloniki, PC, Greece
| | - Dimitra Gialamprinou
- 2nd Neonatal Department and Neonatal Intensive Care Unit (NICU), Aristotle University of Thessaloniki, "Papageorgiou" Hospital, Thessaloniki, PC, Greece
| | - Christos-Georgios Kontovazainitis
- 2nd Neonatal Department and Neonatal Intensive Care Unit (NICU), Aristotle University of Thessaloniki, "Papageorgiou" Hospital, Thessaloniki, PC, Greece
| | - Athanasios Moraitis
- 2nd Neonatal Department and Neonatal Intensive Care Unit (NICU), Aristotle University of Thessaloniki, "Papageorgiou" Hospital, Thessaloniki, PC, Greece
| | - Georgios Katsaras
- 2nd Neonatal Department and Neonatal Intensive Care Unit (NICU), Aristotle University of Thessaloniki, "Papageorgiou" Hospital, Thessaloniki, PC, Greece
| | - Abraham Pouliakis
- 2nd Department of Pathology, National and Kapodistrian University of Athens, "Attikon" University Hospital, Athens, PC, Greece
| | - Elissavet Diamanti
- 2nd Neonatal Department and Neonatal Intensive Care Unit (NICU), Aristotle University of Thessaloniki, "Papageorgiou" Hospital, Thessaloniki, PC, Greece
| |
Collapse
|
4
|
Mansour HM. The interference between SARS-COV-2 and Alzheimer's disease: Potential immunological and neurobiological crosstalk from a kinase perspective reveals a delayed pandemic. Ageing Res Rev 2024; 94:102195. [PMID: 38244862 DOI: 10.1016/j.arr.2024.102195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/22/2024]
Abstract
Coronavirus disease 2019 (COVID-19) has infected over 700 million people, with up to 30% developing neurological manifestations, including dementias. However, there is a lack of understanding of common molecular brain markers causing Alzheimer's disease (AD). COVID-19 has etiological cofactors with AD, making patients with AD a vulnerable population at high risk of experiencing more severe symptoms and worse consequences. Both AD and COVID-19 have upregulated several shared kinases, leading to the repositioning of kinase inhibitors (KIs) for the treatment of both diseases. This review provides an overview of the interactions between the immune system and the nervous system in relation to receptor tyrosine kinases, including epidermal growth factor receptors, vascular growth factor receptors, and non-receptor tyrosine kinases such as Bruton tyrosine kinase, spleen tyrosine kinase, c-ABL, and JAK/STAT. We will discuss the promising results of kinase inhibitors in pre-clinical and clinical studies for both COVID-19 and Alzheimer's disease (AD), as well as the challenges in repositioning KIs for these diseases. Understanding the shared kinases between AD and COVID-19 could help in developing therapeutic approaches for both.
Collapse
Affiliation(s)
- Heba M Mansour
- General Administration of Innovative Products, Central Administration of Biological, Innovative Products, and Clinical Studies (Bio-INN), Egyptian Drug Authority (EDA), Giza, Egypt.
| |
Collapse
|
5
|
Bahk J, Rehman A, Ho KS, Narasimhan B, Baloch HNUA, Zhang J, Yip R, Lookstein R, Steiger DJ. Predictors of pulmonary embolism in hospitalized patients with COVID-19. Thromb J 2023; 21:73. [PMID: 37400813 PMCID: PMC10316556 DOI: 10.1186/s12959-023-00518-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/24/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND High venous thromboembolism (VTE) rates have been described in critically ill patients with COVID-19. We hypothesized that specific clinical characteristics may help differentiate hypoxic COVID-19 patients with and without a diagnosed pulmonary embolism (PE). METHODS We performed a retrospective observational case-control study of 158 consecutive patients hospitalized in one of four Mount Sinai Hospitals with COVID-19 between March 1 and May 8, 2020, who received a Chest CT Pulmonary Angiogram (CTA) to diagnose a PE. We analyzed demographic, clinical, laboratory, radiological, treatment characteristics, and outcomes in COVID-19 patients with and without PE. RESULTS 92 patients were negative (CTA-), and 66 patients were positive for PE (CTA+). CTA + had a longer time from symptom onset to admission (7 days vs. 4 days, p = 0.05), higher admission biomarkers, notably D-dimer (6.87 vs. 1.59, p < 0.0001), troponin (0.015 vs. 0.01, p = 0.01), and peak D-dimer (9.26 vs. 3.8, p = 0.0008). Predictors of PE included time from symptom onset to admission (OR = 1.11, 95% CI 1.03-1.20, p = 0.008), and PESI score at the time of CTA (OR = 1.02, 95% CI 1.01-1.04, p = 0.008). Predictors of mortality included age (HR 1.13, 95% CI 1.04-1.22, p = 0.006), chronic anticoagulation (13.81, 95% CI 1.24-154, p = 0.03), and admission ferritin (1.001, 95% CI 1-1.001, p = 0.01). CONCLUSIONS In 158 hospitalized COVID-19 patients with respiratory failure evaluated for suspected PE, 40.8% patients had a positive CTA. We identified clinical predictors of PE and mortality from PE, which may help with early identification and reduction of PE-related mortality in patients with COVID-19.
Collapse
Affiliation(s)
- Jeeyune Bahk
- Department of Medicine, Mount Sinai Morningside and Mount Sinai West, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Abdul Rehman
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, ISA, USA
| | - Kam Sing Ho
- Department of Medicine, Mount Sinai Morningside and Mount Sinai West, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bharat Narasimhan
- Department of Medicine, Mount Sinai Morningside and Mount Sinai West, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hafiza Noor Ul Ain Baloch
- Division of Pulmonary and Critical Care, Department of Medicine, Mount Sinai West and Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Jiafang Zhang
- Department of Biostatistics, Mount Sinai West and Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rowena Yip
- Department of Biostatistics, Mount Sinai West and Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Lookstein
- Department of Radiology, Mount Sinai Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David J Steiger
- Division of Pulmonary and Critical Care, Department of Medicine, Mount Sinai West and Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA.
| |
Collapse
|
6
|
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
|
7
|
Uzun G, Althaus K, Hammer S, Bakchoul T. Assessment and Monitoring of Coagulation in Patients with COVID-19: A Review of Current Literature. Hamostaseologie 2022; 42:409-419. [PMID: 35477118 DOI: 10.1055/a-1755-8676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Coagulation abnormalities are common in patients with COVID-19 and associated with high morbidity and mortality. It became a daily challenge to navigate through these abnormal laboratory findings and deliver the best possible treatment to the patients. The unique character of COVID-19-induced coagulopathy necessitates not only a dynamic follow-up of the patients in terms of hemostatic findings but also the introduction of new diagnostic methods to determine the overall function of the coagulation system in real time. After the recognition of the high risk of thromboembolism in COVID-19, several professional societies published their recommendations regarding anticoagulation in patients with COVID-19. This review summarizes common hemostatic findings in COVID-19 patients and presents the societal recommendations regarding the use of coagulation laboratory findings in clinical decision-making. Although several studies have investigated coagulation parameters in patients with COVID-19, the methodological shortcomings of published studies as well as the differences in employed anticoagulation regimens that have changed over time, depending on national and international guidelines, limit the applicability of these findings in other clinical settings. Accordingly, evidence-based recommendations for diagnostics during acute COVID-19 infection are still lacking. Future studies should verify the role of coagulation parameters as well as viscoelastic methods in the management of patients with COVID-19.
Collapse
Affiliation(s)
- Günalp Uzun
- Center for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| | - Karina Althaus
- Center for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany.,Medical Faculty of Tuebingen, Institute for Clinical and Experimental Transfusion Medicine, Tuebingen, Germany
| | - Stefanie Hammer
- Center for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| | - Tamam Bakchoul
- Center for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany.,Medical Faculty of Tuebingen, Institute for Clinical and Experimental Transfusion Medicine, Tuebingen, Germany
| |
Collapse
|
8
|
Uzun G, Singh A, Abou-Khalel W, Pelzl L, Weich K, Nowak-Harnau S, Althaus K, Bugert P, Klüter H, Bakchoul T. Platelets and Sera from Donors of Convalescent Plasma after Mild COVID-19 Show No Procoagulant Phenotype. Hamostaseologie 2022; 42:S14-S23. [DOI: 10.1055/a-1797-0564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023] Open
Abstract
AbstractCoronavirus disease-2019 (COVID-19) is associated with increased thromboembolic complications. Long-term alteration in the coagulation system after acute COVID-19 infection is still a subject of research. Furthermore, the effect of sera from convalescent subjects on platelets is not known. In this study, we investigated platelet phenotype, coagulation, and fibrinolysis in COVID-19 convalescent plasma (CCP) donors and analyzed convalescent sera-induced effects on platelets. We investigated CCP donors who had a history of mild COVID-19 infection and donors who did not have COVID-19 were used as controls. We analyzed phosphatidylserine (PS) externalization, CD62p expression, and glycoprotein VI (GPVI) shedding both in platelet-rich plasma (PRP) and after incubation of washed healthy platelets with donors' sera using flow cytometry. Coagulation and fibrinolysis systems were assessed with thromboelastometry. Forty-seven CCP donors (22 males, 25 females; mean age (±SD): 41.4 ± 13.7 years) with a history of mild COVID-19 infection were included. Median duration after acute COVID-19 infection was 97 days (range, 34–401). We did not find an increased PS externalization, CD62p expression, or GPVI shedding in platelets from CCP donors. Sera from CCP donors did not induce PS externalization or GPVI shedding in healthy platelets. Sera-induced CD62p expression was slightly, albeit statistically significantly, lower in CCP donors than in plasma donors without a history of COVID-19. One patient showed increased maximum clot firmness and prolonged lysis time in thromboelastometry. Our findings suggest that procoagulant platelet phenotype is not present after mild COVID-19. Furthermore, CCP sera do not affect the activation status of platelets.
Collapse
Affiliation(s)
- Günalp Uzun
- Centre for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Anurag Singh
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University Hospital of Tübingen, Tübingen, Germany
| | - Wissam Abou-Khalel
- Centre for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Lisann Pelzl
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University Hospital of Tübingen, Tübingen, Germany
| | - Karoline Weich
- Centre for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Stefanie Nowak-Harnau
- Centre for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Karina Althaus
- Centre for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University Hospital of Tübingen, Tübingen, Germany
| | - Peter Bugert
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Hessen, Mannheim, Germany
| | - Harald Klüter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Hessen, Mannheim, Germany
| | - Tamam Bakchoul
- Centre for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University Hospital of Tübingen, Tübingen, Germany
| |
Collapse
|
9
|
Anticoagulation in hospitalized patients with COVID-19. Blood 2022; 140:809-814. [PMID: 35653590 PMCID: PMC9361053 DOI: 10.1182/blood.2021014527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/18/2022] [Indexed: 11/20/2022] Open
Abstract
Coronavirus disease-19 (COVID-19) includes a thromboinflammatory syndrome that may manifest with microvascular and macrovascular thrombosis. Patients with COVID-19 have a higher incidence of venous thromboembolism than other hospitalized patients. Three randomized control trials suggesting benefit of therapeutic heparin in hospitalized noncritically ill patients with COVID-19 have led to conditional guideline recommendations for this treatment. By contrast, prophylactic-dose heparin is recommended for critically ill patients. Unprecedented collaboration and rapidly funded research have improved care of hospitalized patients with COVID-19.
Collapse
|
10
|
Duo H, Li Y, Sun Y, Wei L, Wang Z, Fang F, Zhong Y, Huang J, Luo L, Peng Z, Pan H. Effect of therapeutic versus prophylactic anticoagulation therapy on clinical outcomes in COVID-19 patients: a systematic review with an updated meta-analysis. Thromb J 2022; 20:47. [PMID: 35999599 PMCID: PMC9395810 DOI: 10.1186/s12959-022-00408-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/14/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Previous studies demonstrate a reduced risk of thrombosis and mortality with anticoagulant treatment in patients with COVID-19 than in those without anticoagulation treatment. However, an open question regarding the efficacy and safety of therapeutic anticoagulation (T-AC) versus a lower dose, prophylaxis anticoagulation (P-AC) in COVID-19 patients is still controversial. METHODS We systematically reviewed currently available randomized clinical trials (RCTs) and observational studies (OBs) from January 8, 2019, to January 8, 2022, and compared prophylactic and therapeutic anticoagulant treatment in COVID-19 patients. The primary outcomes were risk of mortality, major bleeding, and the secondary outcomes included venous and arterial thromboembolism. Subgroup analysis was also performed between critically ill and non-critically ill patients with COVID-19 and between patients with higher and lower levels of D-dimer. Sensitivity analysis was performed to decrease the bias and the impact of population heterogeneity. RESULTS We identified 11 RCTs and 17 OBs fulfilling our inclusion criteria. In the RCTs analyses, there was no statistically significant difference in the relative risk of mortality between COVID-19 patients with T-AC treatment and those treated with P-AC (RR 0.95, 95% CI, 0.78-1.15, P = 0.60). Similar results were also found in the OBs analyses (RR 1.21, 95% CI, 0.98-1.49, P = 0.08). The pooling meta-analysis using a random-effects model combined with effect sizes showed that in the RCTs and OBs analyses, patients with COVID-19 who received T-AC treatment had a significantly higher relative risk of the major bleeding event than those with P-AC treatment in COVID-19 patients (RCTs: RR 1.76, 95% CI, 1.19-2.62, P = 0.005; OBs: RR 2.39, 95% CI, 1.56-3.68, P < 0.0001). Compared with P-AC treatment in COVID-19 patients, patients with T-AC treatment significantly reduced the incidence of venous thromboembolism (RR 0.51, 95% CI, 0.39-0.67, P<0.00001), but it is not associated with arterial thrombosis events (RR 0.97, 95% CI, 0.66-1.42, P = 0.87). The subgroup analysis of OBs shows that the mortality risk significantly reduces in critically ill COVID-19 patients treated with T-AC compared with those with P-AC treatment (RR 0.58, 95% CI, 0.39-0.86, P = 0.007), while the mortality risk significantly increases in non-critically ill COVID-19 patients treated with T-AC (RR 1.56, 95% CI, 1.34-1.80, P < 0.00001). In addition, T-AC treatment does not reduce the risk of mortality in COVID-19 patients with high d-dimer levels in RCTs. Finally, the overall sensitivity analysis after excluding two RCTs studies remains consistent with the previous results. CONCLUSIONS In our integrated analysis of included RCTs and OBs, there is no significant difference between the mortality of T-AC and P-AC treatment in unselected patients with COVID-19. T-AC treatment in COVID-19 patients significantly reduced the incidence of venous thromboembolism but showed a higher risk of bleeding than those with P-AC treatment. In addition, P-AC treatment was superior to T-AC treatment in non-critically ill COVID-19 patients, the evidence supporting the necessity for T-AC treatment in critically ill COVID-19 patients came only from OBs. TRIAL REGISTRATION Protocol registration: The protocol was registered at PROSPERO (CRD42021293294).
Collapse
Affiliation(s)
- Hong Duo
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, 169 Eastlake Rd., Wuchang, Wuhan, 430071, Hubei province, China
- The Second Clinical College of Wuhan University, Wuhan, 430071, China
| | - Yahui Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, 169 Eastlake Rd., Wuchang, Wuhan, 430071, Hubei province, China
- The Second Clinical College of Wuhan University, Wuhan, 430071, China
| | - Yujie Sun
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, 169 Eastlake Rd., Wuchang, Wuhan, 430071, Hubei province, China
- The Second Clinical College of Wuhan University, Wuhan, 430071, China
| | - Liang Wei
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, 169 Eastlake Rd., Wuchang, Wuhan, 430071, Hubei province, China
- The Second Clinical College of Wuhan University, Wuhan, 430071, China
| | - Ziqing Wang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, 169 Eastlake Rd., Wuchang, Wuhan, 430071, Hubei province, China
- The Second Clinical College of Wuhan University, Wuhan, 430071, China
| | - Fang Fang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yuxin Zhong
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, 169 Eastlake Rd., Wuchang, Wuhan, 430071, Hubei province, China
- Clinical Research Center for Critical Care Medicine of Hubei Province, Wuhan, 430071, China
| | - Jiao Huang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Linjie Luo
- Department of Experimental Radiation Oncology & Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, 77030, USA.
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, 169 Eastlake Rd., Wuchang, Wuhan, 430071, Hubei province, China.
- Clinical Research Center for Critical Care Medicine of Hubei Province, Wuhan, 430071, China.
| | - Huaqin Pan
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, 169 Eastlake Rd., Wuchang, Wuhan, 430071, Hubei province, China.
- Clinical Research Center for Critical Care Medicine of Hubei Province, Wuhan, 430071, China.
| |
Collapse
|
11
|
Jalali KS, Basala A, Habeb M. Arterio-Venous Thrombosis and Spontaneous Bleeding in COVID-19-Associated Coagulopathy: A Case Report. Cureus 2022; 14:e27770. [PMID: 36106271 PMCID: PMC9450432 DOI: 10.7759/cureus.27770] [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: 08/08/2022] [Indexed: 11/20/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome which was declared a global pandemic in 2019 causing significant morbidities and mortalities. COVID-19 is a multi-systemic disease and is not primarily limited to the respiratory system. Thrombus formation is one of its distinct features. However, renal complications associated with COVID-19 are rarely reported in the literature due to limited occurrence and research. We report a rare case of right retroperitoneal hematoma in a COVID-19 patient. We report a 51-year-old male patient who was received at the emergency department (ED). The patient was positive for COVID-19 and had a Glasgow coma scale of 12/15. The patient was initially managed on IV anticoagulation due to cavernous sinus thrombosis and was placed on mechanical ventilation which helped him to improve. After two weeks, a sudden drop in hemoglobin was observed. CT scan of abdomen and pelvis showed the presence of a right retroperitoneal hematoma, and right renal artery non-occlusive filling defect. The patient was successfully managed with conservative treatment. Retroperitoneal hematoma although a rare occurrence in COVID-19 patient should be observed and monitored closely in case of bleeding or anemia, as early management and intervention is beneficial.
Collapse
|
12
|
Vitaliti G, Giacchi V, Sciacca M, Ruggieri M, Falsaperla R. Thrombotic events in children and adolescent patients with SARS-Cov-2 infection: a systematic review with meta-analysis on incidence and management. Expert Rev Hematol 2022; 15:635-643. [PMID: 35757861 DOI: 10.1080/17474086.2022.2094758] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES the present paper aimed to study available literature on the hypercoagulability state of pediatric patients affected by COVID-19, and the current management of thrombosis in these patients, considering that no guidelines have been published since now in this age group. METHODS N 244 titles were screened using the selected MESH words, 180 abstracts and 120 full texts were read, 12 articles were included, and four were analysed by meta-analysis. RESULTS Over 1128 COVID-19 positive patients, nearly half of them developed inflammatory sequelae, and 7.35% (40 patients over 544 who developed MIS-C) had thrombotic events. Less than 50% of patients with inflammatory disease were under anticoagulant prophylactic treatment, and doses of anticoagulant protocols vary from different centres. Thrombotic events prevented after the start of unfractioned heparin (UFH) therapy, even if 1.06% (4 patients) died. Only two patients presented complications after anticoagulant treatment, with non-fatal bleeding after UFH treatment. No other complications were reported. No difference in the incidence of thrombotic events between patients under prophylactic low molecular weight heparin (LMWH) and without was found in meta-analysis (p=0.32). CONCLUSIONS Little is known on the incidence and management of hyper coagulopathy in pediatric COVID-19 infection. Further studies have to clarify this topic.
Collapse
Affiliation(s)
- Giovanna Vitaliti
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico - San Marco, San Marco Hospital, Via Azeglio Ciampi, 95100, Catania, Italy
| | - Valentina Giacchi
- Neonatal Intensive Care Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico - San Marco, San Marco Hospital, San Marco Hospital, Via Azeglio Ciampi, 95100, Catania, Italy
| | - Monica Sciacca
- Post graduate program in Pediatric, Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - Martino Ruggieri
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, 95124, Catania, Italy
| | - Raffaele Falsaperla
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico - San Marco, San Marco Hospital, Via Azeglio Ciampi, 95100, Catania, Italy.,Neonatal Intensive Care Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico - San Marco, San Marco Hospital, San Marco Hospital, Via Azeglio Ciampi, 95100, Catania, Italy
| |
Collapse
|
13
|
Wang LG, Wang L. Current Strategies in Treating Cytokine Release Syndrome Triggered by Coronavirus SARS-CoV-2. Immunotargets Ther 2022; 11:23-35. [PMID: 35611161 PMCID: PMC9124488 DOI: 10.2147/itt.s360151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/07/2022] [Indexed: 12/15/2022] Open
Abstract
Since the beginning of the SARS-CoV-2 pandemic, the treatments and management of the deadly COVID-19 disease have made great progress. The strategies for developing novel treatments against COVID-19 include antiviral small molecule drugs, cell and gene therapies, immunomodulators, neutralizing antibodies, and combination therapies. Among them, immunomodulators are the most studied treatments. The small molecule antiviral drugs and immunoregulators are expected to be effective against viral variants of SARS-CoV-2 as these drugs target either conservative parts of the virus or common pathways of inflammation. Although the immunoregulators have shown benefits in reducing mortality of cytokine release syndrome (CRS) triggered by SARS-CoV-2 infections, extensive investigations on this class of treatment to launch novel therapies that substantially improve efficacy and reduce side effects are still warranted. Moreover, great challenges have emerged as the SARS-CoV-2 virus quickly, frequently, and continuously evolved. This review provides an update and summarizes the recent advances in the treatment of COVID-19 and in particular emphasized the strategies in managing CRS triggered by SARS-CoV-2. A brief perspective in the battle against the deadly disease was also provided.
Collapse
Affiliation(s)
- Long G Wang
- Department of Research and Development, Natrogen Therapeutics International, Inc., Valhalla, NY, USA
| | - Luxi Wang
- Department of Clinical Research, Clinipace Clinical Research, Morrisville, NC, USA
| |
Collapse
|
14
|
Čulić V, Vio R, Proietti R. Thromboprophylaxis for COVID-19-related coagulopathy: what next? EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2022; 8:E18-E19. [PMID: 35104876 PMCID: PMC9383404 DOI: 10.1093/ehjcvp/pvac009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/29/2022] [Indexed: 11/14/2022]
Affiliation(s)
- Viktor Čulić
- Department of Cardiology and Angiology, University Hospital Centre Split, Šoltanska 1, 21000 Split, Croatia
- Department of Clinical Propedeutics, University of Split School of Medicine, Split, Croatia
| | - Riccardo Vio
- Cardiology Unit, Department of Cardiothoracic and Vascular Medicine, Ospedale dell'Angelo, Venice, Italy
| | - Riccardo Proietti
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK
| |
Collapse
|
15
|
Abstract
SARS-CoV-2 virus has become a global health problem that has caused millions of deaths worldwide. The infection can present with multiple clinical features ranging from asymptomatic or mildly symptomatic patients to patients with severe or critical illness that can even lead to death. Although the immune system plays an important role in pathogen control, SARS-CoV-2 can drive dysregulation of this response and trigger severe immunopathology. Exploring the mechanisms of the immune response involved in host defense against SARS-CoV-2 allows us to understand its immunopathogenesis and possibly detect features that can be used as potential therapies to eliminate the virus. The main objective of this review on SARS-CoV-2 is to highlight the interaction between the virus and the immune response. We explore the function and action of the immune system, the expression of molecules at the site of infection that cause hyperinflammation and hypercoagulation disorders, the factors leading to the development of pneumonia and subsequent severe acute respiratory distress syndrome which is the leading cause of death in patients with COVID-19.
Collapse
Affiliation(s)
- Dennis Jiménez
- Departamento de Ciencias de la Vida y Agricultura, Carrera de Ingeniería en Biotecnología, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Pichincha, Ecuador
| | - Marbel Torres Arias
- Departamento de Ciencias de la Vida y Agricultura, Carrera de Ingeniería en Biotecnología, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Pichincha, Ecuador.,Laboratorio de Inmunología y Virología, CENCINAT, GISAH, Universidad de las Fuerzas Armadas, Sangolquí, Pichincha, Ecuador
| |
Collapse
|
16
|
Kaur S, Singh A, Kaur J, Verma N, Pandey AK, Das S, Bhattacharyya S, Guchhait P. Upregulation of cytokine signalling in platelets increases risk of thrombophilia in severe COVID-19 patients. Blood Cells Mol Dis 2022; 94:102653. [PMID: 35180460 PMCID: PMC8832951 DOI: 10.1016/j.bcmd.2022.102653] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 01/08/2023]
Abstract
Abnormal coagulation dynamics, including disseminated intravascular coagulopathy, pulmonary embolism, venous thromboembolism and risk of thrombosis are often associated with the severity of COVID-19. However, very little is known about the contribution of platelets in above pathogenesis. In order to decipher the pathophysiology of thrombophilia in COVID-19, we recruited severely ill patients from ICU, based on the above symptoms and higher D-dimer levels, and compared these parameters with their asymptomatic counterparts. Elevated levels of platelet-derived microparticles and platelet-leukocyte aggregates suggested the hyperactivation of platelets in ICU patients. Strikingly, platelet transcriptome analysis showed a greater association of IL-6 and TNF signalling pathways in ICU patients along with higher plasma levels of IL-6 and TNFα. In addition, upregulation of pathways like blood coagulation and hemostasis, as well as inflammation coexisted in platelets of these patients. Further, the increment of necrotic pathway and ROS-metabolic processes in platelets was suggestive of its procoagulant phenotype in ICU patients. This study suggests that higher plasma IL-6 and TNFα may trigger platelet activation and coagulation, and in turn aggravate thrombosis and hypercoagulation in severe COVID-19 patients. Therefore, the elevated IL-6 and TNFα, may serve as potential risk factors for platelet activation and thrombophilia in these patients.
Collapse
Affiliation(s)
- Simrandeep Kaur
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Anamika Singh
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Jaskaran Kaur
- Translational Health Science Technology Institute, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Nikhil Verma
- ESIC Medical College and Hospital, Faridabad, India
| | | | - Suman Das
- ESIC Medical College and Hospital, Faridabad, India
| | - Sankar Bhattacharyya
- Translational Health Science Technology Institute, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India.
| |
Collapse
|
17
|
Ahmad R, Haque M. Surviving the Storm: Cytokine Biosignature in SARS-CoV-2 Severity Prediction. Vaccines (Basel) 2022; 10:vaccines10040614. [PMID: 35455363 PMCID: PMC9026643 DOI: 10.3390/vaccines10040614] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The world has been stricken mentally, physically, and economically by the COVID-19 virus. However, while SARS-CoV-2 viral infection results in mild flu-like symptoms in most patients, a number of those infected develop severe illness. These patients require hospitalization and intensive care. The severe disease can spiral downwards with eventual severe damage to the lungs and failure of multiple organs, leading to the individual’s demise. It is necessary to identify those who are developing a severe form of illness to provide early management. Therefore, it is crucial to learn about the mechanisms and chemical mediators that lead to critical conditions in SARS-CoV-2 infection. This paper reviews studies regarding the individual chemical mediators, pathways, and means that contribute to worsening health conditions in SARS-CoV-2 infection. Abstract A significant part of the world population has been affected by the devastating SARS-CoV-2 infection. It has deleterious effects on mental and physical health and global economic conditions. Evidence suggests that the pathogenesis of SARS-CoV-2 infection may result in immunopathology such as neutrophilia, lymphopenia, decreased response of type I interferon, monocyte, and macrophage dysregulation. Even though most individuals infected with the SARS-CoV-2 virus suffer mild symptoms similar to flu, severe illness develops in some cases, including dysfunction of multiple organs. Excessive production of different inflammatory cytokines leads to a cytokine storm in COVID-19 infection. The large quantities of inflammatory cytokines trigger several inflammation pathways through tissue cell and immune cell receptors. Such mechanisms eventually lead to complications such as acute respiratory distress syndrome, intravascular coagulation, capillary leak syndrome, failure of multiple organs, and, in severe cases, death. Thus, to devise an effective management plan for SARS-CoV-2 infection, it is necessary to comprehend the start and pathways of signaling for the SARS-CoV-2 infection-induced cytokine storm. This article discusses the current findings of SARS-CoV-2 related to immunopathology, the different paths of signaling and other cytokines that result in a cytokine storm, and biomarkers that can act as early signs of warning for severe illness. A detailed understanding of the cytokine storm may aid in the development of effective means for controlling the disease’s immunopathology. In addition, noting the biomarkers and pathophysiology of severe SARS-CoV-2 infection as early warning signs can help prevent severe complications.
Collapse
Affiliation(s)
- Rahnuma Ahmad
- Department of Physiology, Medical College for Women and Hospital, Plot No 4 Road 8/9, Sector-1, Dhaka 1230, Bangladesh;
| | - Mainul Haque
- Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sungai Besi, Kuala Lumpur 57000, Malaysia
- Correspondence: or
| |
Collapse
|
18
|
Sridharan M, D Navitskas S, M Kock E, E Houghton D, M Heikal N, Chen D, K Pruthi R. Evaluation of soluble fibrin monomer complex in patients in SARS-CoV-2 COVID-19 infection-associated coagulopathy. Eur J Haematol 2022; 108:319-326. [PMID: 34921683 DOI: 10.1111/ejh.13738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 11/29/2022]
Abstract
Hospitalized patients with COVID-19 infection frequently have coagulopathy resembling disseminated intravascular coagulation (DIC). An elevation of D-dimer level is associated with a poor prognosis; however, the role of other fibrin degradation products, such as soluble fibrin monomers (SFMC), is not known. The objective of the study was to investigate the frequency and prognostic role of elevated SFMC in patients with COVID-19. In this retrospective cohort study, patients hospitalized between April 1, 2020 and December 14, 2020 at Mayo Clinic with COVID-19 infection who underwent DIC panel testing were identified. Results of laboratory tests and outcomes (thrombosis and death) within 40 days of testing were obtained via medical record review. Of 108 patients, D-dimer was elevated in 82 (75.9%) patients. Of those with elevated D-dimer, SFMC was elevated in 19/82 (23%) patients. There were 16 thrombotic events and 16 deaths during the 40-day follow-up. The incidence of overt-DIC was 4.6%. In univariate analysis, D-dimer ≥5 x highest upper limit normal (ULN) and elevated SFMC were each associated with higher 40-day mortality. However, when used in combination with D-dimer ≥5 x highest ULN, an elevated SFMC provided no further mortality predictive value. Compared to 75.9% of patients with elevated D-dimers, of those tested, only 23% had elevated SFMC. These results support the hypothesis that elevated D-dimer in COVID-19 infection is a direct consequence of endothelial damage and not overt-DIC.
Collapse
Affiliation(s)
- Meera Sridharan
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Serena D Navitskas
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth M Kock
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Damon E Houghton
- Division of Cardiovascular Disease, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Nahla M Heikal
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Dong Chen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Rajiv K Pruthi
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
19
|
Long B, Gottlieb M. Higher-dose versus standard-dose prophylactic anticoagulation in hospitalized patients with COVID-19. Acad Emerg Med 2022; 29:123-124. [PMID: 34779548 PMCID: PMC8652652 DOI: 10.1111/acem.14417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Brit Long
- Department of Emergency Medicine Brooke Army Medical Center Fort Sam Houston Texas USA
| | - Michael Gottlieb
- Department of Emergency Medicine Rush University Medical Center Chicago Illinois USA
| |
Collapse
|
20
|
OUP accepted manuscript. Lab Med 2022; 53:349-359. [DOI: 10.1093/labmed/lmac005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
21
|
Poveda-Jaramillo R. Coronavirus disease 2019-induced hypercoagulability and its clinical implications. Asian Cardiovasc Thorac Ann 2021; 30:515-523. [PMID: 34930050 DOI: 10.1177/02184923211069185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 is the disease produced by severe acute respiratory syndrome-coronavirus-2, which is introduced into the host's cell thanks to the angiotensin-converting enzyme 2 receptor. Once there, it uses the cell's machinery to multiply itself. In this process, it generates an immune response that stimulates the lymphocytes to produce cytokines and reactive oxygen species that begin to deteriorate the endothelial cell. Complement activation, through the complement attack complex and C5a, contributes to this endothelial damage. The different mediators further promote the expression of adhesion molecules on the endothelial surface, which encourages all blood cells to adhere to the endothelial surface to form small conglomerates, called clots, which obstruct the lumen of the small blood vessels. Furthermore, the mediators of clot lysis are inhibited. All this promotes a prothrombotic environment within the pulmonary capillaries that is reflected in the elevation of D-dimer. The only solution for this cascade of events seems to be the implementation of an effective anticoagulation protocol that early counteracts the changes induced by thrombi in the pulmonary circulation and reflected in the functioning of the right ventricle.
Collapse
|
22
|
Mizurini DM, Hottz ED, Bozza PT, Monteiro RQ. Fundamentals in Covid-19-Associated Thrombosis: Molecular and Cellular Aspects. Front Cardiovasc Med 2021; 8:785738. [PMID: 34977191 PMCID: PMC8718518 DOI: 10.3389/fcvm.2021.785738] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/23/2021] [Indexed: 01/08/2023] Open
Abstract
The novel coronavirus disease (COVID-19) is associated with a high incidence of coagulopathy and venous thromboembolism that may contribute to the worsening of the clinical outcome in affected patients. Marked increased D-dimer levels are the most common laboratory finding and have been repeatedly reported in critically ill COVID-19 patients. The infection caused by Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is followed by a massive release of pro-inflammatory cytokines, which mediate the activation of endothelial cells, platelets, monocytes, and neutrophils in the vasculature. In this context, COVID-19-associated thrombosis is a complex process that seems to engage vascular cells along with soluble plasma factors, including the coagulation cascade, and complement system that contribute to the establishment of the prothrombotic state. In this review, we summarize the main findings concerning the cellular mechanisms proposed for the establishment of COVID-19-associated thrombosis.
Collapse
Affiliation(s)
- Daniella M. Mizurini
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Eugenio D. Hottz
- Oswaldo Cruz Foundation, Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
- Laboratory of Immunothrombosis, Department of Biochemistry, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Brazil
| | - Patrícia T. Bozza
- Oswaldo Cruz Foundation, Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Robson Q. Monteiro
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Gerber GF, Chaturvedi S. How to recognize and manage COVID-19-associated coagulopathy. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2021; 2021:614-620. [PMID: 34889412 PMCID: PMC8791093 DOI: 10.1182/hematology.2021000297] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
COVID-19 is frequently associated with abnormalities on coagulation testing and a coagulopathy driven by inflammation, intravascular coagulation activation, and microvascular thrombosis. Elevated D-dimer is the most common finding and is a predictor of adverse outcomes including thrombosis, critical illness, and death. Although COVID-19-associated coagulopathy has some similarities to disseminated intravascular coagulation, the platelet count is usually preserved, coagulation times are usually normal or minimally prolonged, and thrombosis is more common than bleeding, at least in noncritically ill patients. Bleeding is uncommon but may be a significant problem in critically ill patients, including those who may develop a consumptive coagulopathy with frank disseminated intravascular coagulation and those on extracorporeal membrane oxygenation. Blood product support to correct coagulopathy is reserved for bleeding patients or those requiring invasive procedures. Current recommendations suggest that all hospitalized patients should receive at least a prophylactic dose of anticoagulation. Results from a multiplatform randomized clinical trial suggest that therapeutically dosed anticoagulation may improve outcomes, including the need for organ support and mortality in moderately ill patients but not in those requiring critical care. The results of ongoing trials evaluating the impact of different antithrombotic strategies (therapeutic agents and intensity) on COVID-19 outcomes are eagerly awaited and are expected to have important implications for patient management. We also discuss COVID-19 vaccine-associated cytopenias and bleeding as well as vaccine-induced thrombotic thrombocytopenia, in which thrombosis is associated with thrombocytopenia, elevated D-dimer, and, frequently, hypofibrinogenemia.
Collapse
Affiliation(s)
- Gloria F Gerber
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Shruti Chaturvedi
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
24
|
D’Agnillo F, Walters KA, Xiao Y, Sheng ZM, Scherler K, Park J, Gygli S, Rosas LA, Sadtler K, Kalish H, Blatti CA, Zhu R, Gatzke L, Bushell C, Memoli MJ, O’Day SJ, Fischer TD, Hammond TC, Lee RC, Cash JC, Powers ME, O’Keefe GE, Butnor KJ, Rapkiewicz AV, Travis WD, Layne SP, Kash JC, Taubenberger JK. Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19. Sci Transl Med 2021; 13:eabj7790. [PMID: 34648357 PMCID: PMC11000440 DOI: 10.1126/scitranslmed.abj7790] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is characterized by respiratory distress, multiorgan dysfunction, and, in some cases, death. The pathological mechanisms underlying COVID-19 respiratory distress and the interplay with aggravating risk factors have not been fully defined. Lung autopsy samples from 18 patients with fatal COVID-19, with symptom onset-to-death times ranging from 3 to 47 days, and antemortem plasma samples from 6 of these cases were evaluated using deep sequencing of SARS-CoV-2 RNA, multiplex plasma protein measurements, and pulmonary gene expression and imaging analyses. Prominent histopathological features in this case series included progressive diffuse alveolar damage with excessive thrombosis and late-onset pulmonary tissue and vascular remodeling. Acute damage at the alveolar-capillary barrier was characterized by the loss of surfactant protein expression with injury to alveolar epithelial cells, endothelial cells, respiratory epithelial basal cells, and defective tissue repair processes. Other key findings included impaired clot fibrinolysis with increased concentrations of plasma and lung plasminogen activator inhibitor-1 and modulation of cellular senescence markers, including p21 and sirtuin-1, in both lung epithelial and endothelial cells. Together, these findings further define the molecular pathological features underlying the pulmonary response to SARS-CoV-2 infection and provide important insights into signaling pathways that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Felice D’Agnillo
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | | | - Yongli Xiao
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zong-Mei Sheng
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Jaekeun Park
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sebastian Gygli
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Luz Angela Rosas
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kaitlyn Sadtler
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Heather Kalish
- Bioengineering and Physical Sciences Shared Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Charles A. Blatti
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ruoqing Zhu
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Lisa Gatzke
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Colleen Bushell
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Matthew J. Memoli
- Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Raymond C. Lee
- Division of Cardiothoracic Surgery, USC Keck School of Medicine, Los Angeles, CA, USA
| | - J. Christian Cash
- Division of Cardiothoracic Surgery, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Matthew E. Powers
- Division of Cardiothoracic Surgery, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Grant E. O’Keefe
- Department of Surgery, University of Washington, Harborview Medical Center, Seattle, WA, USA
| | - Kelly J. Butnor
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT, USA
| | - Amy V. Rapkiewicz
- Department of Pathology, New York University Long Island School of Medicine, Mineola, NY, USA
| | - William D. Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - John C. Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffery K. Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
25
|
Lorini FL, Di Matteo M, Gritti P, Grazioli L, Benigni A, Zacchetti L, Bianchi I, Fabretti F, Longhi L. Coagulopathy and COVID-19. Eur Heart J Suppl 2021; 23:E95-E98. [PMID: 34650364 PMCID: PMC8503535 DOI: 10.1093/eurheartj/suab100] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
SARS-CoV-2 infection is associated with frequent thrombotic events, at the micro and macro-vascular level, due to the perpetuation of a state of hypercoagulability. The so-called ‘COVID-19 associated coagulopathy’ (CAC) represents a key aspect in the genesis of organ damage from SARS-CoV-2. The main coagulative alterations described in the literature are represented by high levels of D-dimer and fibrinogen. Although CAC has some common features with disseminated intravascular coagulation and sepsis-induced coagulopathy, there are important differences between these clinical pictures and the phenotype of CAC is unique. The pathogenesis of CAC is complex and is affected by the strong interconnection between the inflammatory system and coagulation, in the phenomenon of immunothrombosis and thrombo-inflammation. Several mechanisms come into play, such as inflammatory cytokines, neutrophils, the complement system as well as an alteration of the fibrinolytic system. Finally, an altered platelet function and especially endothelial dysfunction also play a central role in the pathophysiology of CAC. Heparin has several potential effects in CAC, in fact in addition to the anticoagulant effect, it could have a direct antiviral effect and anti-inflammatory properties. The high incidence of thrombo-embolic phenomena despite the use of antithrombotic prophylaxis have led some experts to recommend the use of anticoagulant doses of heparin, but at present the optimal anticoagulant regimen remains to be determined.
Collapse
Affiliation(s)
- Ferdinando Luca Lorini
- Dipartimento di Anestesia e Terapia Intensiva, Ospedale Papa Giovanni XXIII, Bergamo, Italia
| | - Maria Di Matteo
- Dipartimento di Anestesia e Terapia Intensiva, Ospedale Papa Giovanni XXIII, Bergamo, Italia
| | - Paolo Gritti
- Dipartimento di Anestesia e Terapia Intensiva, Ospedale Papa Giovanni XXIII, Bergamo, Italia
| | - Lorenzo Grazioli
- Dipartimento di Anestesia e Terapia Intensiva, Ospedale Papa Giovanni XXIII, Bergamo, Italia
| | - Alberto Benigni
- Dipartimento di Anestesia e Terapia Intensiva, Ospedale Papa Giovanni XXIII, Bergamo, Italia
| | - Lucia Zacchetti
- Dipartimento di Anestesia e Terapia Intensiva, Ospedale Papa Giovanni XXIII, Bergamo, Italia
| | - Isabella Bianchi
- Dipartimento di Anestesia e Terapia Intensiva, Ospedale Papa Giovanni XXIII, Bergamo, Italia
| | - Fabrizio Fabretti
- Dipartimento di Anestesia e Terapia Intensiva, Ospedale Papa Giovanni XXIII, Bergamo, Italia
| | - Luca Longhi
- Dipartimento di Anestesia e Terapia Intensiva, Ospedale Papa Giovanni XXIII, Bergamo, Italia
| |
Collapse
|
26
|
Jandial A, Gupta A, Malviya A, Agastam S, Kumar D. Coagulation abnormalities & thromboprophylaxis in COVID-19. Indian J Med Res 2021; 153:606-618. [PMID: 34643567 PMCID: PMC8555598 DOI: 10.4103/ijmr.ijmr_3841_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The ongoing pandemic of novel coronavirus 2019 is rapidly evolving, and newer organ- and system-specific manifestations are being observed. Thrombotic complications and coagulopathy are frequent manifestations of the disease, especially in sick patients, which appear to be unique and distinct from sepsis-induced coagulopathy, disseminated intravascular coagulation and other viral infection-induced coagulation abnormalities. Elevated D-dimers and fibrinogen in the early stage of the disease with minimally deranged prothrombin time and platelet counts are prominent and distinguishing features. Venous and arterial thromboses, as opposed to bleeding events, are the major clinical correlates. There is much to be known about the pathogenesis of COVID-associated coagulopathy; however, the mechanisms overlap with thrombotic microangiopathy, haemophagocytic syndrome and antiphospholipid syndrome compounded by the diffuse endothelial damage. The recommendations regarding the treatment are still evolving, but antithrombotic therapy has a definite role in positive outcomes of sick patients.
Collapse
Affiliation(s)
- Aditya Jandial
- Department of Hematology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Anunay Gupta
- Department of Cardiology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Amit Malviya
- Department of Cardiology, North Eastern Indira Gandhi Regional Institute of Health & Medical Sciences, Shillong, Meghalaya, India
| | - Sourabh Agastam
- Department of Cardiology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Dilip Kumar
- Department of Cardiology, Cardiac Catheterization Laboratory, Medical Institute of Cardiovascular Sciences, Kolkata, West Bengal, India
| |
Collapse
|
27
|
Sholzberg M, Tang GH, Rahhal H, AlHamzah M, Kreuziger LB, Áinle FN, Alomran F, Alayed K, Alsheef M, AlSumait F, Pompilio CE, Sperlich C, Tangri S, Tang T, Jaksa P, Suryanarayan D, Almarshoodi M, Castellucci LA, James PD, Lillicrap D, Carrier M, Beckett A, Colovos C, Jayakar J, Arsenault MP, Wu C, Doyon K, Andreou ER, Dounaevskaia V, Tseng EK, Lim G, Fralick M, Middeldorp S, Lee AYY, Zuo F, da Costa BR, Thorpe KE, Negri EM, Cushman M, Jüni P. Effectiveness of therapeutic heparin versus prophylactic heparin on death, mechanical ventilation, or intensive care unit admission in moderately ill patients with covid-19 admitted to hospital: RAPID randomised clinical trial. BMJ 2021; 375:n2400. [PMID: 34649864 PMCID: PMC8515466 DOI: 10.1136/bmj.n2400] [Citation(s) in RCA: 238] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To evaluate the effects of therapeutic heparin compared with prophylactic heparin among moderately ill patients with covid-19 admitted to hospital wards. DESIGN Randomised controlled, adaptive, open label clinical trial. SETTING 28 hospitals in Brazil, Canada, Ireland, Saudi Arabia, United Arab Emirates, and US. PARTICIPANTS 465 adults admitted to hospital wards with covid-19 and increased D-dimer levels were recruited between 29 May 2020 and 12 April 2021 and were randomly assigned to therapeutic dose heparin (n=228) or prophylactic dose heparin (n=237). INTERVENTIONS Therapeutic dose or prophylactic dose heparin (low molecular weight or unfractionated heparin), to be continued until hospital discharge, day 28, or death. MAIN OUTCOME MEASURES The primary outcome was a composite of death, invasive mechanical ventilation, non-invasive mechanical ventilation, or admission to an intensive care unit, assessed up to 28 days. The secondary outcomes included all cause death, the composite of all cause death or any mechanical ventilation, and venous thromboembolism. Safety outcomes included major bleeding. Outcomes were blindly adjudicated. RESULTS The mean age of participants was 60 years; 264 (56.8%) were men and the mean body mass index was 30.3 kg/m2. At 28 days, the primary composite outcome had occurred in 37/228 patients (16.2%) assigned to therapeutic heparin and 52/237 (21.9%) assigned to prophylactic heparin (odds ratio 0.69, 95% confidence interval 0.43 to 1.10; P=0.12). Deaths occurred in four patients (1.8%) assigned to therapeutic heparin and 18 patients (7.6%) assigned to prophylactic heparin (0.22, 0.07 to 0.65; P=0.006). The composite of all cause death or any mechanical ventilation occurred in 23 patients (10.1%) assigned to therapeutic heparin and 38 (16.0%) assigned to prophylactic heparin (0.59, 0.34 to 1.02; P=0.06). Venous thromboembolism occurred in two patients (0.9%) assigned to therapeutic heparin and six (2.5%) assigned to prophylactic heparin (0.34, 0.07 to 1.71; P=0.19). Major bleeding occurred in two patients (0.9%) assigned to therapeutic heparin and four (1.7%) assigned to prophylactic heparin (0.52, 0.09 to 2.85; P=0.69). CONCLUSIONS In moderately ill patients with covid-19 and increased D-dimer levels admitted to hospital wards, therapeutic heparin was not significantly associated with a reduction in the primary outcome but the odds of death at 28 days was decreased. The risk of major bleeding appeared low in this trial. TRIAL REGISTRATION ClinicalTrials.gov NCT04362085.
Collapse
Affiliation(s)
- Michelle Sholzberg
- Departments of Medicine, and Laboratory Medicine and Pathobiology, St Michael's Hospital, Li Ka Shing Knowledge Institute, University of Toronto, Toronto, ON, Canada
| | - Grace H Tang
- Haematology-Oncology Clinical Research Group, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Hassan Rahhal
- Internal Medicine Department, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), University of São Paulo, São Paulo, Brazil
| | - Musaad AlHamzah
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Division of Vascular Surgery, King Saud University Medical City, Riyadh, Saudi Arabia
| | | | - Fionnuala Ní Áinle
- Mater Misericordiae University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- Irish Network for Venous Thromboembolism Research, Dublin, Ireland
| | - Faris Alomran
- Department of Vascular Surgery, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khalid Alayed
- Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Alsheef
- Medical Specialties Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Fahad AlSumait
- Medical Specialties Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Carlos Eduardo Pompilio
- Internal Medicine Department, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), University of São Paulo, São Paulo, Brazil
| | - Catherine Sperlich
- Charles-Lemoyne Hospital, CISSS of Montérégie-Centre, University of Sherbrooke, Greenfield Park, QC, Canada
| | - Sabrena Tangri
- Department of Medicine, William Osler Health System, Brampton, ON, Canada
| | - Terence Tang
- Institute for Better Health, Trillium Health Partners, Department of Medicine, University of Toronto, ON, Canada
| | - Peter Jaksa
- St Joseph's Health Centre, Unity Health Toronto, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Deepa Suryanarayan
- Department of Medicine, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | | | - Lana A Castellucci
- Department of Medicine, Ottawa Hospital Research Institute at the University of Ottawa, Ottawa, ON, Canada
| | - Paula D James
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Marc Carrier
- Department of Medicine, Ottawa Hospital Research Institute at the University of Ottawa, Ottawa, ON, Canada
| | - Andrew Beckett
- St Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Canadian Forces Health Services, Ottawa, Ontario, Canada
| | - Christos Colovos
- Division of Acute Care Surgery, Department of Surgery, University of Vermont Medical Center, VT, USA
- University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Jai Jayakar
- Southlake Regional Health Centre, University of Toronto, Newmarket, ON, Canada
| | | | - Cynthia Wu
- Division of Haematology, University of Alberta Hospital, University of Alberta, Edmonton, AB, Canada
| | - Karine Doyon
- Hospital of the Sacred Heart of Montreal, University of Montréal, Montréal, QC, Canada
| | | | - Vera Dounaevskaia
- Department of Medicine, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Eric K Tseng
- St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Gloria Lim
- St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Michael Fralick
- General Internal Medicine, Sinai Health, University of Toronto, Toronto, ON, Canada
| | - Saskia Middeldorp
- Department of Internal Medicine and Radboud Institute of Health Sciences, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Agnes Y Y Lee
- Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Fei Zuo
- Applied Health Research Centre, St Michael's Hospital, Li Ka Shing Knowledge Institute, University of Toronto, Toronto, ON, Canada
| | - Bruno R da Costa
- Applied Health Research Centre, St Michael's Hospital, Li Ka Shing Knowledge Institute, University of Toronto, Toronto, ON, Canada
- Institute of Primary Health Care, University of Bern, Bern, Switzerland
| | - Kevin E Thorpe
- Applied Health Research Centre, St Michael's Hospital, Li Ka Shing Knowledge Institute, University of Toronto, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Elnara Márcia Negri
- Laboratory of Medical Investigation (LIM-59), Cellular Biology, Department of Pathology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Mary Cushman
- Departments of Medicine and Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, University of Vermont Medical Center, Burlington, VT, USA
| | - Peter Jüni
- Applied Health Research Centre, St Michael's Hospital, Li Ka Shing Knowledge Institute, University of Toronto, Toronto, ON, Canada
- Departments of Medicine and Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
28
|
Global haemostatic tests demonstrate the absence of parameters of hypercoagulability in non-hypoxic mild COVID-19 patients: a prospective matched study. J Thromb Thrombolysis 2021; 53:646-662. [PMID: 34581945 PMCID: PMC8476716 DOI: 10.1007/s11239-021-02575-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/12/2021] [Indexed: 01/22/2023]
Abstract
Severe COVID-19 patients demonstrate hypercoagulability, necessitating thromboprophylaxis. However, less is known about the haemostatic profile in mild COVID-19 patients. We performed an age and gender-matched prospective study of 10 severe and 10 mild COVID-19 patients. Comprehensive coagulation profiling together with Thromboelastography and Clot Waveform Analysis were performed. FBC, PT, APTT, D-dimer, fibrinogen and CWA were repeated every 3 days for both groups and repeat TEG was performed for severe patients up till 15 days. On recruitment, severe patients had markers reflecting hypercoagulability including raised median D-dimer 1.0 μg/mL (IQR 0.6, 1.4) (p = 0.0004), fibrinogen 5.6 g/L (IQR 4.9, 6.6) (p = 0.002), Factor VIII 206% (IQR 171, 203) and vWF levels 265.5% (IQR 206, 321). Mild patients had normal values of PT, aPTT, fibrinogen and D-dimer, and slightly elevated median Factor VIII and von Willebrand factor (vWF) levels. Repeated 3-day assessments for both groups showed declining trends in D-dimer and Fibrinogen. CWA of severe COVID-19 group demonstrated hypercoagulability with an elevated median values of aPTT delta change 78.8% (IQR 69.8, 85.2) (p = 0.001), aPTT clot velocity (min1) 7.8%/s (IQR 6.7, 8.3) (p = 0.001), PT delta change 22.4% (IQR 19.4, 29.5) (p = 0.004), PT min1 7.1%/s (IQR 6.3, 9.0) (p = 0.02), PT clot acceleration (min 2) 3.6%/s2 (IQR 3.2, 4.5) (p = 0.02) and PT clot deceleration (max2) 2.9%/s2 (IQR 2.5, 3.5) (p = 0.02). TEG of severe patients reflected hypercoagulability with significant increases in the median values of CFF MA 34.6 mm (IQR 27.4,38.6) (p = 0.003), CRT Angle 78.9° (IQR 78.3, 80.0) (p = 0.0006), CRT A10 67.6 mm (IQR 65.8, 69.6) (p = 0.007) and CFF A10 32.0 mm (IQR 26.8, 34.0) (p = 0.003). Mild COVID-19 patients had absent hypercoagulability in both CWA and TEG. 2 severe patients developed thromboembolic events while none occurred in the mild COVID-19 group. Mild COVID-19 patients show absent parameters of hypercoagulability in global haemostatic tests while those with severe COVID-19 demonstrated parameters associated with hypercoagulability on the global haemostatic tests together with raised D-Dimer, fibrinogen, Factor VIII and vWF levels.
Collapse
|
29
|
Philadelphia-Negative Chronic Myeloproliferative Neoplasms during the COVID-19 Pandemic: Challenges and Future Scenarios. Cancers (Basel) 2021; 13:cancers13194750. [PMID: 34638236 PMCID: PMC8507529 DOI: 10.3390/cancers13194750] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/09/2021] [Accepted: 09/17/2021] [Indexed: 12/30/2022] Open
Abstract
An outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) started in December 2019 in China and then become pandemic in February 2020. Several publications investigated the possible increased rate of COVID-19 infection in hematological malignancies. Based on the published data, strategies for the management of chronic Philadelphia-negative chronic myeloproliferative neoplasms (MPNs) are provided. The risk of severe COVID-19 seems high in MPN, particularly in patients with essential thrombocythemia, but not negligible in myelofibrosis. MPN patients are at high risk of both thrombotic and hemorrhagic complications and this must be accounted in the case of COVID-19 deciding on a case-by-case basis. There are currently no data to suggest that hydroxyurea or interferon may influence the risk or severity of COVID-19 infection. Conversely, while the immunosuppressive activity of ruxolitinib might pose increased risk of infection, its abrupt discontinuation during COVID-19 syndrome is associated with worse outcome. All MPN patients should receive vaccine against COVID-19; reassuring data are available on efficacy of mRNA vaccines in MPNs.
Collapse
|
30
|
Ortega-Paz L, Galli M, Capodanno D, Franchi F, Rollini F, Bikdeli B, Mehran R, Montalescot G, Gibson CM, Lopes RD, Andreotti F, Angiolillo DJ. Safety and efficacy of different prophylactic anticoagulation dosing regimens in critically and non-critically ill patients with COVID-19: A systematic review and meta-analysis of randomized controlled trials. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2021; 8:677-686. [PMID: 34519777 PMCID: PMC8499924 DOI: 10.1093/ehjcvp/pvab070] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/25/2021] [Accepted: 09/11/2021] [Indexed: 02/06/2023]
Abstract
Background The clinical impact of different prophylactic anticoagulation regimens among hospitalized patients with coronavirus disease 2019 (COVID-19) remains unclear. We pooled evidence from available randomized controlled trials (RCTs) to provide insights on this topic. Methods and results We searched for RCTs comparing treatment with an escalated-dose (intermediate-dose or therapeutic-dose) vs. a standard-dose prophylactic anticoagulation regimen in critically and non-critically ill COVID-19 patients requiring hospitalization and without a formal indication for anticoagulation. The primary efficacy endpoint was all-cause death, and the primary safety endpoint was major bleeding. Seven RCTs were identified, including 5154 patients followed on an average of 33 days. Compared to standard-dose prophylactic anticoagulation, escalated-dose prophylactic anticoagulation was not associated with a reduction of all-cause death [17.8% vs. 18.6%; risk ratio (RR) 0.96, 95% confidence interval (CI) 0.78–1.18] but was associated with an increase in major bleeding (2.4% vs. 1.4%; RR 1.73, 95%CI 1.15–2.60). Compared to prophylactic anticoagulation used at a standard dose, an escalated dose was associated with lower rates of venous thromboembolism (2.5% vs. 4.7%; RR 0.55, 95%CI 0.41–0.74) without a significant effect on myocardial infarction (RR 0.80, 95%CI 0.47–1.36), stroke (RR 0.94, 95%CI 0.43–2.09), or systemic arterial embolism (RR 1.20, 95%CI 0.29–4.95). There were no significant interactions in the subgroup analysis for critically and non-critically ill patients. Conclusions Our findings provide comprehensive and high-quality evidence for the use of standard-dose prophylactic anticoagulation over an escalated-dose regimen as routine standard of care for hospitalized patients with COVID-19 who do not have an indication for therapeutic anticoagulation, irrespective of disease severity. Study registration This study is registered in PROSPERO (CRD42021257203).
Collapse
Affiliation(s)
- Luis Ortega-Paz
- Cardiovascular Institute, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Mattia Galli
- Cardiovascular Medicine, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy.,Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Davide Capodanno
- Division of Cardiology, A.O.U. "Policlinico-Vittorio Emanuele," University of Catania, Catania, Italy
| | - Francesco Franchi
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Fabiana Rollini
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Behnood Bikdeli
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Center for Outcomes Research and Evaluation (CORE), Yale School of Medicine, New Haven, CT, USA; Cardiovascular Research Foundation (CRF), New York, NY, USA
| | - Roxana Mehran
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Gilles Montalescot
- Sorbonne Université, ACTION Study Group, Institut de Cardiologie (Assistance Publique - Hôpitaux de Paris) Hôpital Pitié-Salpêtrière, INSERM UMRS 1166, Paris France
| | - C Michael Gibson
- Cardiovascular Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Renato D Lopes
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, USA; Brazilian Clinical Research Institute, São Paulo, Brazil
| | - Felicita Andreotti
- Cardiovascular Medicine, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| |
Collapse
|
31
|
Saleh M, Alkofide A, Alshammari A, Siddiqui K, Owaidah T. Changes in Hematological, Clinical and Laboratory Parameters for Children with COVID-19: Single-Center Experience. J Blood Med 2021; 12:819-826. [PMID: 34512062 PMCID: PMC8427837 DOI: 10.2147/jbm.s321372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/06/2021] [Indexed: 12/23/2022] Open
Abstract
Background COVID-19 wreaked havoc on the healthcare system, with more than 36 million cases reported globally. Although the pediatric population makes up a lesser proportion of total COVID-19 patients than adults, the clinical status, age and comorbidities warrant identifying possible prognostic factors associated with disease severity in this group. The current study aimed to explore the incidence of thrombosis, overall outcome, and different hematological and coagulation markers in children with COVID-19. Methods This is a single-center prospective study of 43 patients (age < 14 years) with confirmed COVID-19 diagnosis recruited from April to August 2020. Data for clinical presentation were collected and analyzed. The samples were tested for different hematological and coagulation markers. Results Twenty-nine (67.4%) were symptomatic at presentation, with fever being the most common symptom (n = 23, 53.5%), followed by respiratory (n = 5, 11.6%) and gastrointestinal symptoms (n = 3, 7%). Co-morbid conditions were recorded in 26 (60.5%) patients, with malignancy being the commonest (n = 9, 20.9%). In this cohort of patients with age <14 years, hypertension, respiratory symptoms and ABO group-A were significantly associated with pediatric intensive care unit (PICU) admission during the course of treatment. Patients with elevated FVIII and fibrinogen levels at presentation were more likely to have an extended length of hospital stay (LOS) (P-value =0.036 and 0.032 respectively). No thrombotic event was observed in our cohort. D-dimer values were higher (above 0.5 µg/mL) in 24 (55.8%) patients at admission. We found an association between high D-dimer and PICU admission and LOS. Conclusion Although we did not observe thrombosis in our cohort, serial measurements of D-dimer and elevated FVIII bear a prognostic value in predicting the need for critical care in children with COVID-19. Further studies with larger sample size can aid in the establishment of prognostic factors for the pediatric COVID-19 population.
Collapse
Affiliation(s)
- Mahasen Saleh
- Department of Pediatric Hematology Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Amani Alkofide
- Department of Pediatric Hematology Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Anfal Alshammari
- Department of Pediatric Hematology Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Khawar Siddiqui
- Department of Pediatric Hematology Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Tarek Owaidah
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Kingdom of Saudi Arabia.,Department of Pathology, Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| |
Collapse
|
32
|
Sholzberg M, Tang GH, Rahhal H, AlHamzah M, Kreuziger LB, Ní Áinle F, Alomran F, Alayed K, Alsheef M, AlSumait F, Pompilio CE, Sperlich C, Tangri S, Tang T, Jaksa P, Suryanarayan D, Almarshoodi M, Castellucci L, James PD, Lillicrap D, Carrier M, Beckett A, Colovos C, Jayakar J, Arsenault MP, Wu C, Doyon K, Andreou ER, Dounaevskaia V, Tseng EK, Lim G, Fralick M, Middeldorp S, Lee AYY, Zuo F, da Costa BR, Thorpe KE, Negri EM, Cushman M, Jüni P. Heparin for Moderately Ill Patients with Covid-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 34268513 DOI: 10.1101/2021.07.08.21259351] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background Heparin, in addition to its anticoagulant properties, has anti-inflammatory and potential anti-viral effects, and may improve endothelial function in patients with Covid-19. Early initiation of therapeutic heparin could decrease the thrombo-inflammatory process, and reduce the risk of critical illness or death. Methods We randomly assigned moderately ill hospitalized ward patients admitted for Covid-19 with elevated D-dimer level to therapeutic or prophylactic heparin. The primary outcome was a composite of death, invasive mechanical ventilation, non-invasive mechanical ventilation or ICU admission. Safety outcomes included major bleeding. Analysis was by intention-to-treat. Results At 28 days, the primary composite outcome occurred in 37 of 228 patients (16.2%) assigned to therapeutic heparin, and 52 of 237 patients (21.9%) assigned to prophylactic heparin (odds ratio, 0.69; 95% confidence interval [CI], 0.43 to 1.10; p=0.12). Four patients (1.8%) assigned to therapeutic heparin died compared with 18 patients (7.6%) assigned to prophylactic heparin (odds ratio, 0.22; 95%-CI, 0.07 to 0.65). The composite of all-cause mortality or any mechanical ventilation occurred in 23 (10.1%) in the therapeutic heparin group and 38 (16.0%) in the prophylactic heparin group (odds ratio, 0.59; 95%-CI, 0.34 to 1.02). Major bleeding occurred in 2 patients (0.9%) with therapeutic heparin and 4 patients (1.7%) with prophylactic heparin (odds ratio, 0.52; 95%-CI, 0.09 to 2.85). Conclusions In moderately ill ward patients with Covid-19 and elevated D-dimer level, therapeutic heparin did not significantly reduce the primary outcome but decreased the odds of death at 28 days. Trial registration numbers: NCT04362085 ; NCT04444700.
Collapse
|
33
|
Mitrovic M, Sabljic N, Cvetkovic Z, Pantic N, Zivkovic Dakic A, Bukumiric Z, Libek V, Savic N, Milenkovic B, Virijevic M, Vucinic V, Milosevic I, Pravdic Z, Suvajdzic N, Fareed J, Antic D. Rotational thromboelastometry (ROTEM) profiling of COVID-19 patients. Platelets 2021; 32:690-696. [PMID: 33561381 DOI: 10.1080/09537104.2021.1881949] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/22/2020] [Accepted: 01/07/2021] [Indexed: 01/06/2023]
Abstract
We evaluated coagulation abnormalities via traditional tests and rotational thromboelastometry (ROTEM) in a group of 94 patients with confirmed SARS-CoV-2 infection and different severity of pneumonia (34 moderate, 25 severe, 35 critical) with the hypothesis that ROTEM parameters differed by coronavirus disease 2019 (COVID-19) severity. Shorter than normal clotting time (CT) and higher than normal maximum clot firmness (MCF) in extrinsic rotational thromboelastometry (EXTEM) and fibrinogen rotational thromboelastometry (FIBTEM), shorter than normal EXTEM clot formation time (CFT), and higher than normal α-angle were classified as markers of hypercoagulable state. Increment in the number of patients with ≥2 hypercoagulable parameters, higher EXTEM (P = .0001), FIBTEM MCF (P = .0001) and maximum lysis decrement (P = .002) with increment in disease severity was observed (P = .0001). Significant positive correlations between IL6 and CT EXTEM (P = .003), MCF EXTEM (P = .033), MCF FIBTEM (P = .01), and negative with ML EXTEM (P = .006) were seen. Our findings based on analysis of different disease severity groups confirmed that a hypercoagulable ROTEM pattern characterized by clot formation acceleration, high clot strength, and reduced fibrinolysis was more frequent in advanced disease groups and patients with high IL6. These results supported the need for different thromboprophylaxis approaches for different severity groups.
Collapse
Affiliation(s)
- Mirjana Mitrovic
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nikica Sabljic
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia
| | - Zorica Cvetkovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinical Hospital Center Zemun, Zemun, , Serbia
| | - Nikola Pantic
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia
| | | | - Zoran Bukumiric
- Institute for Medical Statistics and Informatics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vesna Libek
- Clinical Hospital Center Zemun, Zemun, , Serbia
| | - Nebojsa Savic
- Clinic of Vascular Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Branislava Milenkovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Pulmology, Clinical Center of Serbia, Belgrade, Serbia
| | - Marijana Virijevic
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Violeta Vucinic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Pulmology, Clinical Center of Serbia, Belgrade, Serbia
| | - Ivana Milosevic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic for Infectious and Tropical Disease, Clinical Center of Serbia, Belgrade, Serbia
| | - Zlatko Pravdic
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia
| | - Nada Suvajdzic
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jawed Fareed
- Departments of Pathology and Pharmacology, Loyola University Center, Maywood, Illinois, USA
| | - Darko Antic
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
34
|
Zanframundo G, Graziani A, Barbara C, Francesco P, Teresa MM, Cristian C, Pierluigi G, Ludovico D. Resolution of pulmonary artery thrombosis in patients with moderate COVID-19 disease. J Community Hosp Intern Med Perspect 2021; 11:470-472. [PMID: 34211650 PMCID: PMC8221146 DOI: 10.1080/20009666.2021.1921908] [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: 12/15/2022] Open
Abstract
Novel Coronavirus disease (COVID-19) has been widely described as the cause for a proinflammatory and hypercoagulable state. The inflammatory process involving the alveolar vascular endothelium in the respiratory system, is a determining factor for the onset of primary Pulmonary Artery Thrombosis (PAT) even in patients with heparin prophylactic treatment. Little is known about the efficacy of the anticoagulant therapy during the course of PAT caused by COVID-19. In this paper we describe the results obtained in patients with moderate COVID-19 disease, previously threated with prophylactic enoxaparin, who then received full Anticoagulant treatment after diagnosis of PAT. After three months Computed Tomography Pulmonary Angiography demonstrated a complete resolution of the vascular obstructive lesions in all patients, while all the coagulation tests were normal.
Collapse
Affiliation(s)
- Grazia Zanframundo
- Department of Internal Medicine, S. Maria delle Croci Hospital Ravenna, Italy
| | - Alessandro Graziani
- Department of Internal Medicine, S. Maria delle Croci Hospital Ravenna, Italy
| | - Caroli Barbara
- Department of Internal Medicine, S. Maria delle Croci Hospital Ravenna, Italy
| | - Palmese Francesco
- Department of Internal Medicine, S. Maria delle Croci Hospital Ravenna, Italy
| | | | | | | | | |
Collapse
|
35
|
Smythe MA, Burns C, Liu Q, Garwood CL. Potential Dexamethasone-Direct Oral Anticoagulant Drug Interaction: Is This a Concern in COVID? Ann Pharmacother 2021; 56:319-329. [PMID: 34137279 DOI: 10.1177/10600280211025042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE To evaluate the literature on a potential dexamethasone-direct oral anticoagulant (DOAC) drug interaction and provide management considerations with COVID hypercoagulability. DATA SOURCES A search of EMBASE, PubMed, and Google Scholar (January 1990 to May 2021), limited to the English language, using applicable search terms resulted in 137 articles, with 21 relevant articles included. Regulatory agency and clinical guidance documents were also reviewed. STUDY SELECTION AND DATA EXTRACTION Included articles describe in vitro or in vivo animal or human data for dexamethasone induction of cytochrome P450 (CYP) 3A4 or P-glycoprotein (P-gp). DATA SYNTHESIS Dexamethasone has the potential to interact with the DOACs via CYP3A4 and/or P-gp induction. Only apixaban and rivaroxaban have CYP3A4 metabolism. Dexamethasone can increase CYP3A4 activity by up to 70% and reduce the area under the concentration-time curve (AUC) of CYP3A4 substrates by >40%, which is consistent with criteria for a weak CYP inducer. In rodents, dexamethasone P-gp induction is associated with AUC reductions of 20% to 50%. Human data are lacking. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE Severe COVID-19 infection is associated with hypercoagulability. Although heparins are the preferred anticoagulants for hospitalized COVID-19 patients, DOACs are being utilized. Dexamethasone is recommended for hospitalized COVID-19 patients requiring supplemental oxygen. The concurrent use of dexamethasone and apixaban or rivaroxaban in such patients carries the potential for reduced anticoagulant effect during a state of heightened thrombotic risk. CONCLUSIONS Concurrent use of dexamethasone and apixaban or rivaroxaban in hospitalized COVID-19 patients with laboratory evidence of COVID coagulopathy should be avoided until higher-quality data are available.
Collapse
Affiliation(s)
- Maureen A Smythe
- Wayne State University, Detroit, MI, USA.,Beaumont Hospital, Royal Oak, MI, USA
| | | | | | - Candice L Garwood
- Wayne State University, Detroit, MI, USA.,Detroit Medical Center, MI, USA
| |
Collapse
|
36
|
Bonnemain J, Ltaief Z, Liaudet L. The Right Ventricle in COVID-19. J Clin Med 2021; 10:jcm10122535. [PMID: 34200990 PMCID: PMC8230058 DOI: 10.3390/jcm10122535] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022] Open
Abstract
Infection with the novel severe acute respiratory coronavirus-2 (SARS-CoV2) results in COVID-19, a disease primarily affecting the respiratory system to provoke a spectrum of clinical manifestations, the most severe being acute respiratory distress syndrome (ARDS). A significant proportion of COVID-19 patients also develop various cardiac complications, among which dysfunction of the right ventricle (RV) appears particularly common, especially in severe forms of the disease, and which is associated with a dismal prognosis. Echocardiographic studies indeed reveal right ventricular dysfunction in up to 40% of patients, a proportion even greater when the RV is explored with strain imaging echocardiography. The pathophysiological mechanisms of RV dysfunction in COVID-19 include processes increasing the pulmonary vascular hydraulic load and others reducing RV contractility, which precipitate the acute uncoupling of the RV with the pulmonary circulation. Understanding these mechanisms provides the fundamental basis for the adequate therapeutic management of RV dysfunction, which incorporates protective mechanical ventilation, the prevention and treatment of pulmonary vasoconstriction and thrombotic complications, as well as the appropriate management of RV preload and contractility. This comprehensive review provides a detailed update of the evidence of RV dysfunction in COVID-19, its pathophysiological mechanisms, and its therapy.
Collapse
Affiliation(s)
- Jean Bonnemain
- Department of Adult Intensive Care Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland; (J.B.); (Z.L.)
| | - Zied Ltaief
- Department of Adult Intensive Care Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland; (J.B.); (Z.L.)
| | - Lucas Liaudet
- Department of Adult Intensive Care Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland; (J.B.); (Z.L.)
- Division of Pathophysiology, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
- Correspondence: ; Tel.: +41-79-556-4278
| |
Collapse
|
37
|
Ford DJ, Duggan NM, Fry SE, Ripoll-Rozada J, Agten SM, Liu W, Corcilius L, Hackeng TM, van Oerle R, Spronk HMH, Ashhurst AS, Mini Sasi V, Kaczmarski JA, Jackson CJ, Pereira PJB, Passioura T, Suga H, Payne RJ. Potent Cyclic Peptide Inhibitors of FXIIa Discovered by mRNA Display with Genetic Code Reprogramming. J Med Chem 2021; 64:7853-7876. [PMID: 34044534 DOI: 10.1021/acs.jmedchem.1c00651] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The contact system comprises a series of serine proteases that mediate procoagulant and proinflammatory activities via the intrinsic pathway of coagulation and the kallikrein-kinin system, respectively. Inhibition of Factor XIIa (FXIIa), an initiator of the contact system, has been demonstrated to lead to thrombo-protection and anti-inflammatory effects in animal models and serves as a potentially safer target for the development of antithrombotics. Herein, we describe the use of the Randomised Nonstandard Peptide Integrated Discovery (RaPID) mRNA display technology to identify a series of potent and selective cyclic peptide inhibitors of FXIIa. Cyclic peptides were evaluated in vitro, and three lead compounds exhibited significant prolongation of aPTT, a reduction in thrombin generation, and an inhibition of bradykinin formation. We also describe our efforts to identify the critical residues for binding FXIIa through alanine scanning, analogue generation, and via in silico methods to predict the binding mode of our lead cyclic peptide inhibitors.
Collapse
Affiliation(s)
- Daniel J Ford
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Nisharnthi M Duggan
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Sarah E Fry
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Jorge Ripoll-Rozada
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Stijn M Agten
- Department of Biochemistry, University of Maastricht, Cardiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Wenyu Liu
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Tokyo 113-0033, Japan
| | - Leo Corcilius
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Tilman M Hackeng
- Department of Biochemistry, University of Maastricht, Cardiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Rene van Oerle
- Department of Biochemistry, University of Maastricht, Cardiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Henri M H Spronk
- Department of Biochemistry, University of Maastricht, Cardiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Anneliese S Ashhurst
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales 2006, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Vishnu Mini Sasi
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra, ACT 0200, Australia.,Research School of Chemistry, Australian National University, Canberra, ACT 0200, Australia
| | - Joe A Kaczmarski
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra, ACT 0200, Australia.,Research School of Chemistry, Australian National University, Canberra, ACT 0200, Australia
| | - Colin J Jackson
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra, ACT 0200, Australia.,Research School of Chemistry, Australian National University, Canberra, ACT 0200, Australia
| | - Pedro José Barbosa Pereira
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Toby Passioura
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia.,Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Tokyo 113-0033, Japan.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia.,Sydney Analytical, The University of Sydney, Sydney, NSW 2006, Australia
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Tokyo 113-0033, Japan
| | - Richard J Payne
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
38
|
Thachil J. Lessons learnt from COVID-19 coagulopathy. ACTA ACUST UNITED AC 2021; 2:577-584. [PMID: 34226900 PMCID: PMC8242569 DOI: 10.1002/jha2.228] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 01/08/2023]
Abstract
The coronavirus disease 2019 (COVID‐19) pandemic has already left an indelible mark in human lives. Despite the havoc it created, this pandemic also saw significant advances in the management of an infectious disease wherein worldwide collaborative efforts from health care professionals have been unprecedented. One of the commonest complications recognised early in the pandemic is the development of coagulopathy. In this review, the lessons learnt from COVID‐19 coagulopathy are summarised with some perspectives on future clinical and research strategies. These include how local versus systemic coagulopathy can matter, how we can put D‐dimers to effective use, exhort more input into identifying a simple platelet activation marker, rethink the role of fibrinogen, look differently at lupus anticoagulant and heparin‐induced thrombocytopenia, bring back disseminated intravascular coagulation into our differential diagnosis slate and most importantly channel more funding into haemostasis and thrombosis research.
Collapse
Affiliation(s)
- Jecko Thachil
- Department of Haematology Manchester University Hospitals Manchester UK
| |
Collapse
|
39
|
Kamel H. We stand ready … Blood collection organizations and the COVID-19 pandemic. Transfusion 2021; 61:1345-1349. [PMID: 33966274 PMCID: PMC8207090 DOI: 10.1111/trf.16400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 04/04/2021] [Indexed: 12/14/2022]
Abstract
See article on page 1471–1478, in this issue
Collapse
|
40
|
Short SAP, Gupta S, Brenner SK, Hayek SS, Srivastava A, Shaefi S, Singh H, Wu B, Bagchi A, Al-Samkari H, Dy R, Wilkinson K, Zakai NA, Leaf DE. d-dimer and Death in Critically Ill Patients With Coronavirus Disease 2019. Crit Care Med 2021; 49:e500-e511. [PMID: 33591017 PMCID: PMC8275993 DOI: 10.1097/ccm.0000000000004917] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Hypercoagulability may be a key mechanism for acute organ injury and death in patients with severe coronavirus disease 2019, but the relationship between elevated plasma levels of d-dimer, a biomarker of coagulation activation, and mortality has not been rigorously studied. We examined the independent association between d-dimer and death in critically ill patients with coronavirus disease 2019. DESIGN Multicenter cohort study. SETTING ICUs at 68 hospitals across the United States. PATIENTS Critically ill adults with coronavirus disease 2019 admitted to ICUs between March 4, 2020, and May 25, 2020, with a measured d-dimer concentration on ICU day 1 or 2. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS The primary exposure was the highest normalized d-dimer level (assessed in four categories: < 2×, 2-3.9×, 4-7.9×, and ≥ 8× the upper limit of normal) on ICU day 1 or 2. The primary endpoint was 28-day mortality. Multivariable logistic regression was used to adjust for confounders. Among 3,418 patients (63.1% male; median age 62 yr [interquartile range, 52-71 yr]), 3,352 (93.6%) had a d-dimer concentration above the upper limit of normal. A total of 1,180 patients (34.5%) died within 28 days. Patients in the highest compared with lowest d-dimer category had a 3.11-fold higher odds of death (95% CI, 2.56-3.77) in univariate analyses, decreasing to a 1.81-fold increased odds of death (95% CI, 1.43-2.28) after multivariable adjustment for demographics, comorbidities, and illness severity. Further adjustment for therapeutic anticoagulation did not meaningfully attenuate this relationship (odds ratio, 1.73; 95% CI, 1.36-2.19). CONCLUSIONS In a large multicenter cohort study of critically ill patients with coronavirus disease 2019, higher d-dimer levels were independently associated with a greater risk of death.
Collapse
Affiliation(s)
- Samuel A P Short
- Larner College of Medicine, University of Vermont, Burlington, VT
| | - Shruti Gupta
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA
| | - Samantha K Brenner
- Department of Internal Medicine, Hackensack Meridian School of Medicine at Seton Hall, Nutley, NJ
- Department of Internal Medicine, Heart & Vascular Hospital, Hackensack Meridian Health Hackensack University Medical Center, Hackensack, NJ
| | - Salim S Hayek
- Division of Cardiology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Anand Srivastava
- Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Shahzad Shaefi
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | | | - Benjamin Wu
- Division of Pulmonary, Critical Care & Sleep Medicine, NYU Langone Medical Center, New York, NY
| | - Aranya Bagchi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA
| | - Hanny Al-Samkari
- Division of Hematology, Massachusetts General Hospital, Boston, MA
| | - Rajany Dy
- Department of Medicine, University Medical Center of Southern Nevada Hospital, University of Nevada, Las Vegas, NV
| | - Katherine Wilkinson
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT
| | - Neil A Zakai
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
41
|
Leentjens J, van Haaps TF, Wessels PF, Schutgens REG, Middeldorp S. COVID-19-associated coagulopathy and antithrombotic agents-lessons after 1 year. LANCET HAEMATOLOGY 2021; 8:e524-e533. [PMID: 33930350 PMCID: PMC8078884 DOI: 10.1016/s2352-3026(21)00105-8] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/26/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023]
Abstract
COVID-19 is associated with a high incidence of thrombotic complications, which can be explained by the complex and unique interplay between coronaviruses and endothelial cells, the local and systemic inflammatory response, and the coagulation system. Empirically, an intensified dose of thrombosis prophylaxis is being used in patients admitted to hospital with COVID-19 and several guidelines on this topic have been published, although the insufficiency of high quality and direct evidence has led to weak recommendations. In this Viewpoint we summarise the pathophysiology of COVID-19 coagulopathy in the context of patients who are ambulant, admitted to hospital, and critically ill or non-critically ill, and those post-discharge from hospital. We also review data from randomised controlled trials in the past year of antithrombotic therapy in patients who are critically ill. These data provide the first high-quality evidence on optimal use of antithrombotic therapy in patients with COVID-19. Pharmacological thromboprophylaxis is not routinely recommended for patients who are ambulant and post-discharge. A first ever trial in non-critically ill patients who were admitted to hospital has shown that a therapeutic dose of low-molecular-weight heparin might improve clinical outcomes in this population. In critically ill patients, this same treatment does not improve outcomes and prophylactic dose anticoagulant thromboprophylaxis is recommended. In the upcoming months we expect numerous data from the ongoing antithrombotic COVID-19 studies to guide clinicians at different stages of the disease.
Collapse
Affiliation(s)
- Jenneke Leentjens
- Department of Internal Medicine, Radboud Institute for Health Sciences, Radboud University Medical Centre, Nijmegen, Netherlands.
| | - Thijs F van Haaps
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Pieter F Wessels
- Department of Medical Oncology, University of Pretoria, Pretoria, South Africa; Ampath Laboratories, Pretoria, South Africa
| | - Roger E G Schutgens
- Van Creveldkliniek, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands
| | - Saskia Middeldorp
- Department of Internal Medicine, Radboud Institute for Health Sciences, Radboud University Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
42
|
Ghosh K, Ghosh M. Thrombohaemorhhagic balance in coronavirus disease 2019 and its management: a perspective. Blood Coagul Fibrinolysis 2021; 32:167-171. [PMID: 33827111 DOI: 10.1097/mbc.0000000000000993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Coronavirus disease 2019 infection produce a prothrombotic state. This is initiated through multiple pathways and is finally aggravated by cross talks with cytokine storm and neutrophil, platelet, complement activation. All these combine towards the second week of illness to produce thrombosis in the lung capillaries surrounding the alveolus producing characteristic pulmonary dysfunction (PaO2/FiO2 > 300, normal or minimally increased lung compliance and very high d-dimer levels) and a high rate of peripheral venous thrombosis. International and many national guidelines have approached this state in different ways but all emphasized the need for management and prevention of widespread thrombosis. It is felt more aggressive and graded thrombosis prevention and management should be initiated early in the treatment. d-Dimer, neutrophil count, SaO2, fibrinogen levels should be used to control the hypercoagulability. Drugs like statins which have anti-inflammatory action as well as ability to reduce fibrinogen and other clotting factors should be used in the beginning along with antiplatelet drugs and progressively complement activation and neutrophil extracellular traps inhibitors, oral mucopolysaccharides, full-scale anticoagulation along with judicial use of fibrinolysis supporting drugs should be added. In the present review, we have evaluated the various studies and argued the rationality that the anticoagulation in this condition should be initiated early during the infection and should be increased in a graded manner depending on clinical and laboratory progression of the condition until a strong specific antiviral drug for coronavirus disease 2019 infection is available.
Collapse
Affiliation(s)
| | - Malay Ghosh
- Formerly Head of the Department of Haematology, Nil Ratan Sircar Medical College, Kolkata 7000'2, India
| |
Collapse
|
43
|
Khatri P, Agrawal KK, Sharma D, Chhetri P, Neupane A, Piryani RM, Baral PP, Sapkota SR, Banjade A, Chhetri A, Bhandari S, Bharali S. Prevalence of Elevated D-dimer Levels in Confirmed COVID-19 Cases in Intensive Care Unit of a Tertiary Care Centre of Western Nepal. JNMA J Nepal Med Assoc 2021; 59:243-247. [PMID: 34506437 PMCID: PMC8369534 DOI: 10.31729/jnma.6284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Indexed: 01/15/2023] Open
Abstract
Introduction: D-dimer is currently the best available marker for COVID-19 associated hemostatic abnormalities. This study aims to find out the prevelance of elevated D-dimer levels in confirmed COVID-19 cases in intensive care unit of a tertiary care hospital of western Nepal. Methods: This descriptive cross-sectional study was conducted among 95 patients admitted to COVID Intensive Care Unit of a teriary care centre from August 2020 to January 2021 after taking ethical clearence from Institutional Review Committee in order to determine the D-dimer levels in confirmed COVID-19 cases. D-dimer value was measured at the admission and the highest D-dimer value was recorded during the course of hospital stay with the risk of mortality in confirmed COVID-19 cases. The normal range of D-dimer was taken as <0.35 mg/dl as per our hospital laboratory standards. Convenience sampling method was used. Data entry and descriptive analysis were done in Statistical Package for the Social Sciences version 25.0, point estimate at 95% Confidence Interval was calculated along with frequency and proportion for binary data. Results: Out of total 95 cases of COVID-19 included in this study, 25 (89.3%) patients with age ≥65 years and 42 (62.69%) patients aged <65 years had elevated D-dimer on admission. Data showed that 29 (67.4%) patients having elevated D-dimer at admission had mortality. Conclusions: Elevated D-dimer levels was frequently seen in patients admitted in Intensive Care Unit with COVID-19. Our study suggested that measurement of D-dimer may guide in clinical decision making.
Collapse
Affiliation(s)
- Prabin Khatri
- Department of Internal Medicine, Universal College of Medical Sciences, Bhairahawa, Nepal
| | - Krishna Kumar Agrawal
- Department of Internal Medicine, Universal College of Medical Sciences, Bhairahawa, Nepal
| | | | - Pradip Chhetri
- Department of Community Medicine, Universal College of Medical Sciences, Bhairahawa, Nepal
| | - Aryan Neupane
- Universal College of Medical Sciences, Bhairahawa, Nepal
| | - Rano Mal Piryani
- Department of Internal Medicine, Universal College of Medical Sciences, Bhairahawa, Nepal
| | - Pawan Puspa Baral
- Department of Anesthesiology, Universal College of Medical Sciences, Bhairahawa, Nepal
| | - Suman Raj Sapkota
- Department of Internal Medicine, Universal College of Medical Sciences, Bhairahawa, Nepal
| | - Ashish Banjade
- Universal College of Medical Sciences, Bhairahawa, Nepal
| | | | | | - Swaraj Bharali
- Universal College of Medical Sciences, Bhairahawa, Nepal
| |
Collapse
|
44
|
Henrina J, Santosa Putra IC, Cahyadi I, Lawrensia S, Hadi Gunawan HF, Cahyadi A, Franke J, Suciadi LP. Clinical characteristics and outcomes of venous thromboembolism in patients hospitalized for COVID-19: Systematic review and meta-analysis. THROMBOSIS UPDATE 2021; 2:100037. [PMID: 38620815 PMCID: PMC7857988 DOI: 10.1016/j.tru.2021.100037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/19/2022] Open
Abstract
Venous thromboembolism is prevalent in hospitalized COVID-19 patients. Through systematic review and meta-analysis, we have investigated the differences in clinical characteristics and outcome of hospitalized COVID-19 patients with (+) and without (-) venous thromboembolism (VTE). 45 studies with a total of 8859 patients were included in the qualitative synthesis. Subsequently, 38 studies with a total of 7847 patients, were quantitatively analyzed. There was no mortality difference between the VTE (-) and VTE (+) hospitalized COVID-19 patients (RR1.32 (0.97, 1.79); 0.07; I2 64%, p < 0.001). Patients with VTE (+) were more likely to get admitted to the intensive care unit (RR1.77 (1.26, 2.50); p < 0.001; I2 63%, p = 0.03) and mechanically ventilated (RR 2.35 (1.22, 4.53); p = 0.01; I2 88%, p < 0.001). Moreover, male gender (RR 1.19 (1.14,1.24), p < 0.001; I2 0%, p = 0.68), increased the risk of VTE. Regarding patients lab values', VTE (+) was significantly associated with higher white blood cell, neutrophil count, D-Dimer, alanine aminotransferase (ALT), lactate dehydrogenase (LDH), and C-reactive protein (CRP), along with prolonged prothrombin time. On the contrary, VTE (+) was associated with lower albumin and neutrophil-lymphocyte ratio (NLR). This findings provide the initial framework for risk stratification of hospitalized COVID-19 patients with VTE.
Collapse
Affiliation(s)
| | | | | | - Sherly Lawrensia
- School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jl. Pluit Raya No.2, RT.21/RW.8, Penjaringan, Kec. Penjaringan, Kota Jakarta Utara, Daerah Khusus Ibukota Jakarta, 14440, Indonesia
| | | | - Alius Cahyadi
- Department of Internal Medicine, School of Medicine and Health Sciences. Atma Jaya Catholic University of Indonesia/ Atma Jaya Hospital, Jakarta, Indonesia
| | | | | |
Collapse
|
45
|
Leong K, Gaglani B, Khanna AK, McCurdy MT. Novel Diagnostics and Therapeutics in Sepsis. Biomedicines 2021; 9:biomedicines9030311. [PMID: 33803628 PMCID: PMC8003067 DOI: 10.3390/biomedicines9030311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/13/2021] [Accepted: 03/16/2021] [Indexed: 12/11/2022] Open
Abstract
Sepsis management demands early diagnosis and timely treatment that includes source control, antimicrobial therapy, and resuscitation. Currently employed diagnostic tools are ill-equipped to rapidly diagnose sepsis and isolate the offending pathogen, which limits the ability to offer targeted and lowest-toxicity treatment. Cutting edge diagnostics and therapeutics in development may improve time to diagnosis and address two broad management principles: (1) source control by removing the molecular infectious stimulus of sepsis, and (2) attenuation of the pathological immune response allowing the body to heal. This review addresses novel diagnostics and therapeutics and their role in the management of sepsis.
Collapse
Affiliation(s)
- Kieran Leong
- Division of Pulmonary & Critical Care, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Bhavita Gaglani
- Department of Anesthesiology, Section on Critical Care Medicine, Wake Forest University Hospital, Winston-Salem, NC 27157, USA; (B.G.); (A.K.K.)
| | - Ashish K. Khanna
- Department of Anesthesiology, Section on Critical Care Medicine, Wake Forest University Hospital, Winston-Salem, NC 27157, USA; (B.G.); (A.K.K.)
- Department of Outcomes Research, Outcomes Research Consortium, Cleveland, OH 44195, USA
| | - Michael T. McCurdy
- Division of Pulmonary & Critical Care, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Correspondence:
| |
Collapse
|
46
|
Ortega‐Paz L, Capodanno D, Montalescot G, Angiolillo DJ. Coronavirus Disease 2019-Associated Thrombosis and Coagulopathy: Review of the Pathophysiological Characteristics and Implications for Antithrombotic Management. J Am Heart Assoc 2021; 10:e019650. [PMID: 33228447 PMCID: PMC7955431 DOI: 10.1161/jaha.120.019650] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus-2, which has posed a significant threat to global health. Although the infection is frequently asymptomatic or associated with mild symptoms, in a small proportion of patients it can produce an intense inflammatory and prothrombotic state that can lead to acute respiratory distress syndrome, multiple organ failure, and death. Angiotensin-converting enzyme 2, highly expressed in the respiratory system, has been identified as a functional receptor for severe acute respiratory syndrome coronavirus-2. Notably, angiotensin-converting enzyme 2 is also expressed in the cardiovascular system, and there are multiple cardiovascular implications of COVID-19. Cardiovascular risk factors and cardiovascular disease have been associated with severe manifestations and poor prognosis in patients with COVID-19. More important, patients with COVID-19 may have thrombotic and coagulation abnormalities, promoting a hypercoagulable state and resulting in an increased rate of thrombotic and thromboembolic events. This review will describe the pathophysiological characteristics of the cardiovascular involvement following infection by severe acute respiratory syndrome coronavirus-2, with a focus on thrombotic and thromboembolic manifestations and implications for antithrombotic management.
Collapse
Affiliation(s)
- Luis Ortega‐Paz
- Cardiovascular InstituteInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero Universitaria "Policlinico‐Vittorio Emanuele"University of CataniaCataniaItaly
| | - Gilles Montalescot
- ACTION Study GroupInstitut de CardiologieAssistance Publique ‐ Hôpitaux de ParisHôpital Pitié‐SalpêtrièreUniversity Paris 6INSERM UMRS 1166ParisFrance
| | | |
Collapse
|
47
|
Thachil J. D-Dimer and thrombosis in COVID-19. INDIAN JOURNAL OF VASCULAR AND ENDOVASCULAR SURGERY 2021. [DOI: 10.4103/ijves.ijves_67_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
48
|
Dai W, Rao R, Sher A, Tania N, Musante CJ, Allen R. A Prototype QSP Model of the Immune Response to SARS-CoV-2 for Community Development. CPT Pharmacometrics Syst Pharmacol 2021; 10:18-29. [PMID: 33217169 PMCID: PMC7753647 DOI: 10.1002/psp4.12574] [Citation(s) in RCA: 21] [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: 08/13/2020] [Accepted: 11/04/2020] [Indexed: 12/20/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic requires the rapid development of efficacious treatments for patients with life-threatening coronavirus disease 2019 (COVID-19). Quantitative systems pharmacology (QSP) models are mathematical representations of pathophysiology for simulating and predicting the effects of existing or putative therapies. The application of model-based approaches, including QSP, have accelerated the development of some novel therapeutics. Nevertheless, the development of disease-scale mechanistic models can be a slow process, often taking years to be validated and considered mature. Furthermore, emerging data may make any QSP model quickly obsolete. We present a prototype QSP model to facilitate further development by the scientific community. The model accounts for the interactions between viral dynamics, the major host immune response mediators and tissue damage and regeneration. The immune response is determined by viral activation of innate and adaptive immune processes that regulate viral clearance and cell damage. The prototype model captures two physiologically relevant outcomes following infection: a "healthy" immune response that appropriately defends against the virus, and an uncontrolled alveolar inflammatory response that is characteristic of acute respiratory distress syndrome. We aim to significantly shorten the typical QSP model development and validation timeline by encouraging community use, testing, and refinement of this prototype model. It is our expectation that the model will be further advanced in an open science approach (i.e., by multiple contributions toward a validated quantitative platform in an open forum), with the ultimate goal of informing and accelerating the development of safe and effective treatment options for patients.
Collapse
Affiliation(s)
- Wei Dai
- Early Clinical DevelopmentPfizer Worldwide Research, Development and MedicalCambridgeMassachusettsUSA
| | - Rohit Rao
- Early Clinical DevelopmentPfizer Worldwide Research, Development and MedicalCambridgeMassachusettsUSA
| | - Anna Sher
- Early Clinical DevelopmentPfizer Worldwide Research, Development and MedicalCambridgeMassachusettsUSA
| | - Nessy Tania
- Early Clinical DevelopmentPfizer Worldwide Research, Development and MedicalCambridgeMassachusettsUSA
| | - Cynthia J. Musante
- Early Clinical DevelopmentPfizer Worldwide Research, Development and MedicalCambridgeMassachusettsUSA
| | - Richard Allen
- Early Clinical DevelopmentPfizer Worldwide Research, Development and MedicalCambridgeMassachusettsUSA
| |
Collapse
|
49
|
Obi AT, Barnes GD, Napolitano LM, Henke PK, Wakefield TW. Venous thrombosis epidemiology, pathophysiology, and anticoagulant therapies and trials in severe acute respiratory syndrome coronavirus 2 infection. J Vasc Surg Venous Lymphat Disord 2021; 9:23-35. [PMID: 32916371 PMCID: PMC7834652 DOI: 10.1016/j.jvsv.2020.08.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/12/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus confers a risk of significant coagulopathy, with the resulting development of venous thromboembolism (VTE), potentially contributing to the morbidity and mortality. The purpose of the present review was to evaluate the potential mechanisms that contribute to this increased risk of coagulopathy and the role of anticoagulants in treatment. METHODS A literature review of coronavirus disease 2019 (COVID-19) and/or SARS-CoV-2 and cell-mediated inflammation, clinical coagulation abnormalities, hypercoagulability, pulmonary intravascular coagulopathy, and anticoagulation was performed. The National Clinical Trials database was queried for ongoing studies of anticoagulation and/or antithrombotic treatment or the incidence or prevalence of thrombotic events in patients with SARS-CoV-2 infection. RESULTS The reported rate of VTE among critically ill patients infected with SARS-CoV-2 has been 21% to 69%. The phenomenon of breakthrough VTE, or the acute development of VTE despite adequate chemoprophylaxis or treatment dose anticoagulation, has been shown to occur with severe infection. The pathophysiology of overt hypercoagulability and the development of VTE is likely multifactorial, with evidence supporting the role of significant cell-mediated responses, including neutrophils and monocytes/macrophages, endothelialitis, cytokine release syndrome, and dysregulation of fibrinolysis. Collectively, this inflammatory process contributes to the severe pulmonary pathology experienced by patients with COVID-19. As the infection worsens, extreme D-dimer elevations, significant thrombocytopenia, decreasing fibrinogen, and prolongation of prothrombin time and partial thromboplastin time occur, often associated with deep vein thrombosis, in situ pulmonary thrombi, and/or pulmonary embolism. A new phenomenon, termed pulmonary intravascular coagulopathy, has been associated with morbidity in patients with severe infection. Heparin, both unfractionated heparin and low-molecular-weight heparin, have emerged as agents that can address the viral infection, inflammation, and thrombosis in this syndrome. CONCLUSIONS The overwhelming inflammatory response in patients with SARS-CoV-2 infection can lead to a hypercoagulable state, microthrombosis, large vessel thrombosis, and, ultimately, death. Early VTE prophylaxis should be provided to all admitted patients. Therapeutic anticoagulation therapy might be beneficial for critically ill patients and is the focus of 39 ongoing trials. Close monitoring for thrombotic complications is imperative, and, if confirmed, early transition from prophylactic to therapeutic anticoagulation should be instituted. The interplay between inflammation and thrombosis has been shown to be a hallmark of the SARS-CoV-2 viral infection.
Collapse
Affiliation(s)
- Andrea T Obi
- Section of Vascular Surgery, Department of Surgery, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Mich
| | - Geoffrey D Barnes
- Division of Cardiovascular Medicine, Department of Internal Medicine, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Mich
| | - Lena M Napolitano
- Division of Critical Care Surgery, Department of Surgery, University of Michigan, Ann Arbor, Mich
| | - Peter K Henke
- Section of Vascular Surgery, Department of Surgery, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Mich
| | - Thomas W Wakefield
- Section of Vascular Surgery, Department of Surgery, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
50
|
The Pathobiological Basis for Thrombotic Complications in COVID-19: a Review of the Literature. CURRENT PATHOBIOLOGY REPORTS 2021; 9:107-117. [PMID: 34900401 PMCID: PMC8651460 DOI: 10.1007/s40139-021-00228-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/10/2021] [Indexed: 12/11/2022]
Abstract
Purpose of Review COVID-19 has rapidly evolved into a global pandemic infecting over two hundred and forty-four million individuals to date. In addition to the respiratory sequelae and systemic infection that ensues, an alarming number of micro and macrovascular thrombotic complications have been observed. This review examines the current understanding of COVID-19-associated thrombotic complications, potential mechanisms, and pathobiological basis for thromboses development. Recent Findings The endothelium plays a major role in the process due to direct and indirect injury. The immune system also contributes to a pro-thrombotic environment with immune cell dysregulation leading to excessive formation of cytokines, also called cytokine storm, and an eventual promotion of a hypercoagulable environment, known as immunothrombosis. Additionally, neutrophils play an important role by forming neutrophil extracellular traps, which are shown to be pro-thrombotic and further enhanced in COVID-19 patients. A disruption of the fibrinolysis system has also been observed. Summary Multiple pathways likely contribute synergistically to form a pro-thrombotic milieu. A better understanding of these factors and the complex interplay between them will lead to the improvement of diagnostic and therapeutic interventions.
Collapse
|