1
|
Berra S, Parolin D, Suffritti C, Folcia A, Zanichelli A, Gusso L, Cogliati C, Riva A, Gidaro A, Caccia S. Patterns of C1-Inhibitor Plasma Levels and Kinin-Kallikrein System Activation in Relation to COVID-19 Severity. Life (Basel) 2024; 14:1525. [PMID: 39768234 PMCID: PMC11679851 DOI: 10.3390/life14121525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/16/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Although more than four years have passed since the pandemic began, SARS-CoV-2 continues to be of concern. Therefore, research into the underlying mechanisms that contribute to the development of the disease, especially in more severe forms, remains a priority. Sustained activation of the complement (CS), contact (CAS), and fibrinolytic and kinin-kallikrein systems (KKS) has been shown to play a central role in the pathogenesis of the disease. Since the C1 esterase inhibitor (C1-INH) is a potent inhibitor of all these systems, its role in the disease has been investigated, but some issues remained unresolved. METHODS We evaluated the impact of C1-INH and KKS on disease progression in a cohort of 45 COVID-19 patients divided into groups according to disease severity. We measured plasma levels of total and functional C1-INH and its complexes with kallikrein (PKa), reflecting KKS activation and kallikrein spontaneous activity. RESULTS We observed increased total and functional plasma concentrations of C1-INH in COVID-19 patients. A direct correlation (positive Spearman's r) was observed between C1-INH levels, especially functional C1-INH, and the severity of the disease. Moreover, a significant reduction in the ratio of functional over total C1-INH was evident in patients exhibiting mild to intermediate clinical severity but not in critically ill patients. Accordingly, activation of the KKS, assessed as an increase in PKa:C1-INH complexes, was explicitly observed in the mild categories. CONCLUSIONS Our study's findings on the consumption of C1-INH and the activation of the KKS in the less severe stages of COVID-19 but not in the critical stage suggest a potential role for C1-INH in containing disease severity. These results underscore the importance of C1-INH in the early phases of the disease and its potential implications in COVID-19 progression and/or long-term effects.
Collapse
Affiliation(s)
- Silvia Berra
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Department of Internal Medicine, Ospedale Fatebenefratelli, 20121 Milan, Italy
| | - Debora Parolin
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
| | - Chiara Suffritti
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, 20122 Milan, Italy
| | - Andrea Folcia
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Division of Oncology, Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | - Andrea Zanichelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
- Operative Unit of Medicine, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Luca Gusso
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Internal Medicine Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Chiara Cogliati
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Department of Internal Medicine, Ospedale Luigi Sacco, 20157 Milan, Italy
| | - Agostino Riva
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Department of Infectious Diseases, Ospedale Luigi Sacco, 20157 Milan, Italy
| | - Antonio Gidaro
- Department of Internal Medicine, Ospedale Luigi Sacco, 20157 Milan, Italy
| | - Sonia Caccia
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
| |
Collapse
|
2
|
Padilla S, Prado R, Anitua E. An evolutionary history of F12 gene: Emergence, loss, and vulnerability with the environment as a driver. Bioessays 2023; 45:e2300077. [PMID: 37750435 DOI: 10.1002/bies.202300077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/27/2023]
Abstract
In the context of macroevolutionary transitions, environmental changes prompted vertebrates already bearing genetic variations to undergo gradual adaptations resulting in profound anatomical, physiological, and behavioral adaptations. The emergence of new genes led to the genetic variation essential in metazoan evolution, just as was gene loss, both sources of genetic variation resulting in adaptive phenotypic diversity. In this context, F12-coding protein with defense and hemostatic roles emerged some 425 Mya, and it might have contributed in aquatic vertebrates to the transition from water-to-land. Conversely, the F12 loss in marine, air-breathing mammals like cetaceans has been associated with phenotypic adaptations in some terrestrial mammals in their transition to aquatic lifestyle. More recently, the advent of technological innovations in western lifestyle with blood-contacting devices and harmful environmental nanoparticles, has unfolded new roles of FXII. Environment operates as either a positive or a relaxed selective pressure on genes, and consequently genes are selected or lost. FXII, an old dog facing environmental novelties can learn new tricks and teach us new therapeutic avenues.
Collapse
Affiliation(s)
- Sabino Padilla
- BTI-Biotechnology Institute ImasD, Vitoria, Spain
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Roberto Prado
- BTI-Biotechnology Institute ImasD, Vitoria, Spain
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Eduardo Anitua
- BTI-Biotechnology Institute ImasD, Vitoria, Spain
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| |
Collapse
|
3
|
Ragnoli B, Da Re B, Galantino A, Kette S, Salotti A, Malerba M. Interrelationship between COVID-19 and Coagulopathy: Pathophysiological and Clinical Evidence. Int J Mol Sci 2023; 24:ijms24108945. [PMID: 37240292 DOI: 10.3390/ijms24108945] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/14/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Since the first description of COVID-19 infection, among clinical manifestations of the disease, including fever, dyspnea, cough, and fatigue, it was observed a high incidence of thromboembolic events potentially evolving towards acute respiratory distress syndrome (ARDS) and COVID-19-associated-coagulopathy (CAC). The hypercoagulation state is based on an interaction between thrombosis and inflammation. The so-called CAC represents a key aspect in the genesis of organ damage from SARS-CoV-2. The prothrombotic status of COVID-19 can be explained by the increase in coagulation levels of D-dimer, lymphocytes, fibrinogen, interleukin 6 (IL-6), and prothrombin time. Several mechanisms have been hypothesized to explain this hypercoagulable process such as inflammatory cytokine storm, platelet activation, endothelial dysfunction, and stasis for a long time. The purpose of this narrative review is to provide an overview of the current knowledge on the pathogenic mechanisms of coagulopathy that may characterize COVID-19 infection and inform on new areas of research. New vascular therapeutic strategies are also reviewed.
Collapse
Affiliation(s)
| | - Beatrice Da Re
- Respiratory Unit, Sant'Andrea Hospital, 13100 Vercelli, Italy
| | | | - Stefano Kette
- Respiratory Unit, Sant'Andrea Hospital, 13100 Vercelli, Italy
| | - Andrea Salotti
- Respiratory Unit, Sant'Andrea Hospital, 13100 Vercelli, Italy
| | - Mario Malerba
- Respiratory Unit, Sant'Andrea Hospital, 13100 Vercelli, Italy
- Department of Traslational Medicine, University of Eastern Piedmont (UPO), 28100 Novara, Italy
| |
Collapse
|
4
|
Zerangian N, Erabi G, Poudineh M, Monajjem K, Diyanati M, Khanlari M, Khalaji A, Allafi D, Faridzadeh A, Amali A, Alizadeh N, Salimi Y, Ghane Ezabadi S, Abdi A, Hasanabadi Z, ShojaeiBaghini M, Deravi N. Venous thromboembolism in viral diseases: A comprehensive literature review. Health Sci Rep 2023; 6:e1085. [PMID: 36778773 PMCID: PMC9900357 DOI: 10.1002/hsr2.1085] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/25/2022] [Accepted: 01/19/2023] [Indexed: 02/09/2023] Open
Abstract
Venous thromboembolism (VTE) is known to be a common respiratory and/or cardiovascular complication in hospitalized patients with viral infections. Numerous studies have proven human immunodeficiency virus infection to be a prothrombotic condition. An elevated VTE risk has been observed in critically ill H1N1 influenza patients. VTE risk is remarkably higher in patients infected with the Hepatitis C virus in contrast to uninfected subjects. The elevation of D-dimer levels supported the association between Chikungunya and the Zika virus and the rise of clinical VTE risk. Varicella-zoster virus is a risk factor for both cellulitis and the consequent invasive bacterial disease which may take part in thrombotic initiation. Eventually, hospitalized patients infected with the coronavirus disease of 2019 (COVID-19), the cause of the ongoing worldwide pandemic, could mainly suffer from an anomalous risk of coagulation activation with enhanced venous thrombosis events and poor quality clinical course. Although the risk of VTE in nonhospitalized COVID-19 patients is not known yet, there are a large number of guidelines and studies on thromboprophylaxis administration for COVID-19 cases. This study aims to take a detailed look at the effect of viral diseases on VTE, the epidemiology of VTE in viral diseases, and the diagnosis and treatment of VTE.
Collapse
Affiliation(s)
- Nasibeh Zerangian
- Health Education and Health Promotion, Department of Health Education and Health Promotion, School of HealthMashhad University of Medical SciencesMashhadIran
| | - Gisou Erabi
- Student Research CommitteeUrmia University of Medical SciencesUrmiaIran
| | | | - Kosar Monajjem
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Maryam Diyanati
- Student Research CommitteeRafsanjan University of Medical SciencesRafsanjanIran
| | - Maryam Khanlari
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | | | - Diba Allafi
- Student Research CommitteeUrmia University of Medical SciencesUrmiaIran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, School of MedicineMashhad University of Medical SciencesMashhadIran
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
| | - Arian Amali
- Student Research Committee, Paramedical DepartmentIslamic Azad University, Mashhad BranchMashhadIran
| | - Nilufar Alizadeh
- Doctor of Medicine (MD), School of MedicineIran University of Medical SciencesTehranIran
| | - Yasaman Salimi
- Student Research CommitteeKermanshah University of Medical SciencesKermanshahIran
| | - Sajjad Ghane Ezabadi
- Student's Scientific Research Center, School of MedicineTehran University of Medical SciencesTehranIran
| | - Amir Abdi
- Student Research Committee, School of Medicine, Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Zahra Hasanabadi
- Doctor of Medicine (MD), School of MedicineQazvin University of Medical ScienceQazvinIran
| | - Mahdie ShojaeiBaghini
- Medical Informatics Research Center, Institute for Futures Studies in HealthKerman University of Medical SciencesKermanIran
| | - Niloofar Deravi
- Student Research Committee, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
5
|
Yakovleva EV, Zozulya NI. Physiological and pathological role of factor XII. RUSSIAN JOURNAL OF HEMATOLOGY AND TRANSFUSIOLOGY 2022. [DOI: 10.35754/0234-5730-2022-67-4-570-578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Introduction. The most widely accepted notion of the function of blood clotting factor XII (FXII, Hageman factor) is its involvement in the internal blood clotting pathway. However, the biological role of FXII is diverse.Aim – to review the diverse biological functions of FXII.Main findings. FXII is a serine protease. The structure of FXII has a high degree of homology with plasminogen, tissue plasminogen activator and urokinase plasminogen activator. Activated FXII (FXIIa) has five substrates: high-molecular kininogen, precallikrein, FXI, plasminogen, complement proteins (C1s, C1r). FXII provides hemostatic balance by participating in the processes of blood clotting and fibrinolysis. FXII regulates inflammatory and allergic reactions by interacting with the kallikreinkinin system and the complement system. FXII has biological activity in various cells in vivo: endotheliocytes, platelets, monocytes, neutrophils, fi broblasts, dendritic cells, which determines its diverse role in physiological and pathological processes.
Collapse
|
6
|
Heubner L, Greiner M, Vicent O, Beyer-Westendorf J, Tiebel O, Scholz U, Güldner A, Mirus M, Fries D, Koch T, Spieth PM. Predictive ability of viscoelastic testing using ClotPro® for short-term outcome in patients with severe Covid-19 ARDS with or without ECMO therapy: a retrospective study. Thromb J 2022; 20:48. [PMID: 36038895 PMCID: PMC9421107 DOI: 10.1186/s12959-022-00403-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/04/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND SARS-CoV-2 infections are suspected to trigger the coagulation system through various pathways leading to a high incidence of thromboembolic complications, hypercoagulation and impaired fibrinolytic capacity were previously identified as potentially mechanisms. A reliable diagnostic tool for detecting both is still under discussion. This retrospective study is aimed to examine the prognostic relevance of early viscoelastic testing compared to conventional laboratory tests in COVID-19 patients with acute respiratory distress syndrome (ARDS). METHODS All mechanically ventilated patients with COVID-19 related ARDS treated in our intensive care unit (ICU) between January and March 2021 were included in this study. Viscoelastic testing (VET) was performed using the ClotPro® system after admission to our ICU. Prevalence of thromboembolic events was observed by standardized screening for venous and pulmonary thromboembolism using complete compression ultrasound and thoracic computed tomography pulmonary angiography at ICU admission, respectively. We examined associations between the severity of ARDS at admission to our ICU, in-hospital mortality and the incidence of thromboembolic events comparing conventional laboratory analysis and VET. ECMO related coagulopathy was investigated in a subgroup analysis. The data were analyzed using the Mann-Whitney U test. RESULTS Of 55 patients enrolled in this study, 22 patients required treatment with ECMO. Thromboembolic complications occurred in 51% of all patients. Overall hospital mortality was 55%. In patients with thromboembolic complications, signs of reduced fibrinolytic capacity could be detected in the TPA assay with prolonged lysis time, median 460 s (IQR 350-560) vs 359 s (IQR 287-521, p = 0.073). Patients with moderate to severe ARDS at admission to our ICU showed increased maximum clot firmness as a sign of hypercoagulation in the EX-test (70 vs 67 mm, p < 0.05), FIB-test (35 vs 24 mm, p < 0.05) and TPA-test (52 vs 36 mm, p < 0.05) as well as higher values of inflammatory markers (CRP, PCT and IL6). ECMO patients suffered more frequently from bleeding complications (32% vs 15%). CONCLUSION Although, the predictive value for thromboembolic complications or mortality seems limited, point-of-care viscoelastic coagulation testing might be useful in detecting hypercoagulable states and impaired fibrinolysis in critically ill COVID-19 ARDS patients and could be helpful in identifying patients with a potentially very severe course of the disease.
Collapse
Affiliation(s)
- Lars Heubner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital "Carl Gustav Carus", Technische Universität Dresden, Dresden, Germany
| | - Marvin Greiner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital "Carl Gustav Carus", Technische Universität Dresden, Dresden, Germany
| | - Oliver Vicent
- Department of Anesthesiology and Intensive Care Medicine, University Hospital "Carl Gustav Carus", Technische Universität Dresden, Dresden, Germany
| | - Jan Beyer-Westendorf
- Division of Hematology and Hemostasis, Department of Medicine I, Thrombosis Research University Hospital "Carl Gustav Carus", Technische Universität Dresden, Dresden, Germany
| | - Oliver Tiebel
- Institute of Clinical Chemistry, University Hospital "Carl Gustav Carus", Technische Universität Dresden, Dresden, Germany
| | - Ute Scholz
- MVZ Labor Dr. Reising-Ackermann Und Kollegen, Center of Hemostasis, Leipzig, Germany
| | - Andreas Güldner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital "Carl Gustav Carus", Technische Universität Dresden, Dresden, Germany
| | - Martin Mirus
- Department of Anesthesiology and Intensive Care Medicine, University Hospital "Carl Gustav Carus", Technische Universität Dresden, Dresden, Germany
| | - Dietmar Fries
- Department for General and Surgical Critical Care Medicine, Innsbruck Medical University, Innsbruck, Austria
| | - Thea Koch
- Department of Anesthesiology and Intensive Care Medicine, University Hospital "Carl Gustav Carus", Technische Universität Dresden, Dresden, Germany
| | - Peter Markus Spieth
- Department of Anesthesiology and Intensive Care Medicine, University Hospital "Carl Gustav Carus", Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
7
|
Henderson MW, Lima F, Moraes CRP, Ilich A, Huber SC, Barbosa MS, Santos I, Palma AC, Nunes TA, Ulaf RG, Ribeiro LC, Bernardes AF, Bombassaro B, Dertkigil SSJ, Moretti ML, Strickland S, Annichino-Bizzacchi JM, Orsi FA, Mansour E, Velloso LA, Key NS, De Paula EV. Contact and intrinsic coagulation pathways are activated and associated with adverse clinical outcomes in COVID-19. Blood Adv 2022; 6:3367-3377. [PMID: 35235941 PMCID: PMC8893951 DOI: 10.1182/bloodadvances.2021006620] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/19/2022] [Indexed: 12/27/2022] Open
Abstract
Coagulation activation is a prominent feature of severe acute respiratory syndrome coronavirus 2 (COVID-19) infection. Activation of the contact system and intrinsic pathway has increasingly been implicated in the prothrombotic state observed in both sterile and infectious inflammatory conditions. We therefore sought to assess activation of the contact system and intrinsic pathway in individuals with COVID-19 infection. Baseline plasma levels of protease:serpin complexes indicative of activation of the contact and intrinsic pathways were measured in samples from inpatients with COVID-19 and healthy individuals. Cleaved kininogen, a surrogate for bradykinin release, was measured by enzyme-linked immunosorbent assay, and extrinsic pathway activation was assessed by microvesicle tissue factor-mediated factor Xa (FXa; MVTF) generation. Samples were collected within 24 hours of COVID-19 diagnosis. Thirty patients with COVID-19 and 30 age- and sex-matched controls were enrolled. Contact system and intrinsic pathway activation in COVID-19 was demonstrated by increased plasma levels of FXIIa:C1 esterase inhibitor (C1), kallikrein:C1, FXIa:C1, FXIa:α1-antitrypsin, and FIXa:antithrombin (AT). MVTF levels were also increased in patients with COVID-19. Because FIXa:AT levels were associated with both contact/intrinsic pathway complexes and MVTF, activation of FIX likely occurs through both contact/intrinsic and extrinsic pathways. Among the protease:serpin complexes measured, FIXa:AT complexes were uniquely associated with clinical indices of disease severity, specifically total length of hospitalization, length of intensive care unit stay, and extent of lung computed tomography changes. We conclude that the contact/intrinsic pathway may contribute to the pathogenesis of the prothrombotic state in COVID-19. Larger prospective studies are required to confirm whether FIXa:AT complexes are a clinically useful biomarker of adverse clinical outcomes.
Collapse
Affiliation(s)
- Michael W. Henderson
- University of North Carolina (UNC) Blood Research Center, UNC at Chapel Hill, Chapel Hill, NC
| | - Franciele Lima
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Anton Ilich
- University of North Carolina (UNC) Blood Research Center, UNC at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, UNC at Chapel Hill, Chapel Hill, NC
| | | | - Mayck Silva Barbosa
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Andre C. Palma
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Thyago Alves Nunes
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Raisa Gusso Ulaf
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Luciana Costa Ribeiro
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Ana Flavia Bernardes
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Sergio San Juan Dertkigil
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Maria Luiza Moretti
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY; and
| | - Joyce M. Annichino-Bizzacchi
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
- Hematology and Hemotherapy Center, and
| | - Fernanda Andrade Orsi
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Eli Mansour
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Licio A. Velloso
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Nigel S. Key
- University of North Carolina (UNC) Blood Research Center, UNC at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, UNC at Chapel Hill, Chapel Hill, NC
- Department of Pathology and Laboratory Medicine, UNC at Chapel Hill, Chapel Hill, NC
| | - Erich Vinicius De Paula
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
- Hematology and Hemotherapy Center, and
| |
Collapse
|
8
|
van de Veerdonk FL, Giamarellos-Bourboulis E, Pickkers P, Derde L, Leavis H, van Crevel R, Engel JJ, Wiersinga WJ, Vlaar APJ, Shankar-Hari M, van der Poll T, Bonten M, Angus DC, van der Meer JWM, Netea MG. A guide to immunotherapy for COVID-19. Nat Med 2022; 28:39-50. [PMID: 35064248 DOI: 10.1038/s41591-021-01643-9] [Citation(s) in RCA: 225] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022]
Abstract
Immune dysregulation is an important component of the pathophysiology of COVID-19. A large body of literature has reported the effect of immune-based therapies in patients with COVID-19, with some remarkable successes such as the use of steroids or anti-cytokine therapies. However, challenges in clinical decision-making arise from the complexity of the disease phenotypes and patient heterogeneity, as well as the variable quality of evidence from immunotherapy studies. This Review aims to support clinical decision-making by providing an overview of the evidence generated by major clinical trials of host-directed therapy. We discuss patient stratification and propose an algorithm to guide the use of immunotherapy strategies in the clinic. This will not only help guide treatment decisions, but may also help to design future trials that investigate immunotherapy in other severe infections.
Collapse
Affiliation(s)
- Frank L van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| | | | - Peter Pickkers
- Department of Intensive Care Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lennie Derde
- Department of Intensive Care, University Medical Center Utrecht, Utrecht, the Netherlands.,Julius Center for Health Sciences and Primary Care, Utrecht, the Netherlands
| | - Helen Leavis
- Department of Internal Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Job J Engel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - W Joost Wiersinga
- Division of Infectious Diseases, Center for Experimental Molecular Medicine (CEMM), Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Alexander P J Vlaar
- Department of Intensive Care Medicine and Laboratory of Experimental Intensive Care Medicine and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Manu Shankar-Hari
- School of Immunobiology and Microbial Sciences, King's College London, London, UK
| | - Tom van der Poll
- Division of Infectious Diseases, Center for Experimental Molecular Medicine (CEMM), Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Marc Bonten
- Julius Center for Health Sciences and Primary Care, Utrecht, the Netherlands
| | - Derek C Angus
- UPMC and University of Pittsburgh, Pittsburgh, PA, United States
| | - Jos W M van der Meer
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands. .,Department of Immunology and Metabolism, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany.
| |
Collapse
|
9
|
McKenzie A, Roberts A, Malandkar S, Feuersenger H, Panousis C, Pawaskar D. A phase I, first-in-human, randomized dose-escalation study of anti-activated factor XII monoclonal antibody garadacimab. Clin Transl Sci 2021; 15:626-637. [PMID: 34859955 PMCID: PMC8932690 DOI: 10.1111/cts.13180] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 02/05/2023] Open
Abstract
Factor XII (FXII) is the principal initiator of the plasma contact system and has proinflammatory and prothrombotic activities. This single‐center, first‐in‐human phase I study aimed to assess the safety and tolerability of single escalating doses of garadacimab, a monoclonal antibody that specifically inhibits activated FXII (FXIIa), in healthy male volunteers. Volunteers were randomized to eight cohorts, with intravenous (i.v.) doses of 0.1, 0.3, 1, 3, and 10 mg/kg and subcutaneous (s.c.) doses of 1, 3, and 10 mg/kg. Six volunteers in each cohort received garadacimab or placebo in a ratio of 2:1. Follow‐up for safety lasted 85 days after dosing. Blood samples were collected throughout for pharmacokinetic/pharmacodynamic analysis. Forty‐eight volunteers were enrolled: 32 received garadacimab and 16 received placebo. Most volunteers experienced at least one treatment‐emergent adverse event (TEAE), predominantly grade 1. No serious TEAEs, deaths, or TEAEs leading to discontinuation were reported. No volunteers tested positive for garadacimab antidrug antibodies. Garadacimab plasma concentrations increased in a dose‐dependent manner. Sustained inhibition of FXIIa‐mediated kallikrein activity beyond day 28 resulted from 3 and 10 mg/kg garadacimab (i.v. and s.c.). A dose‐dependent increase in activated partial thromboplastin time with no change in prothrombin time was demonstrated. Garadacimab (single‐dose i.v. and s.c.) was well‐tolerated in healthy volunteers. Dose‐dependent increases in plasma concentration and pharmacodynamic effects in relevant kinin and coagulation pathways were observed. These results support the clinical development of garadacimab, including in phase II studies in hereditary angioedema and coronavirus disease 2019 (COVID‐19).
Collapse
|
10
|
Veronez CL, Christiansen SC, Smith TD, Riedl MA, Zuraw BL. COVID-19 and hereditary angioedema: Incidence, outcomes, and mechanistic implications. Allergy Asthma Proc 2021; 42:506-514. [PMID: 34871158 DOI: 10.2500/aap.2021.42.210083] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Patients with hereditary angioedema (HAE) have been postulated to be at increased risk for coronavirus disease 2019 (COVID-19) infection due to inherent dysregulation of the plasma kallikrein-kinin system. Only limited data have been available to explore this hypothesis. Objective: To assess the interrelationship(s) between COVID-19 and HAE. Methods: Self-reported COVID-19 infection, complications, morbidity, and mortality were surveyed by using an online questionnaire. The participants included subjects with HAE with C1 inhibitor (C1INH) deficiency (HAE-C1INH) and subjects with HAE with normal C1-inhibitor (HAE-nl-C1INH), and household controls (normal controls). The impact of HAE medications was examined. Results: A total of 1162 participants who completed the survey were analyzed, including: 695 subjects with HAE-C1INH, 175 subjects with HAE-nl-C1INH, and 292 normal controls. The incidence of reported COVID-19 was not significantly different between the normal controls (9%) and the subjects with HAE-C1INH (11%) but was greater in the subjects with HAE-nl-C1INH (19%; p = 0.006). Obesity was positively correlated with COVID-19 across the overall population (p = 0.012), with a similar but nonsignificant trend in the subjects with HAE-C1INH. Comorbid autoimmune disease was a risk factor for COVID-19 in the subjects with HAE-C1INH (p = 0.047). COVID-19 severity and complications were similar in all the groups. Reported COVID-19 was reduced in the subjects with HAE-C1INH who received prophylactic subcutaneous C1INH (5.6%; p = 0.0371) or on-demand icatibant (7.8%; p = 0.0016). The subjects with HAE-C1INH and not on any HAE medications had an increased risk of COVID-19 compared with the normal controls (24.5%; p = 0.006). Conclusion: The subjects with HAE-C1INH who were not taking HAE medications had a significantly higher rate of reported COVID-19 infection. Subcutaneous C1INH and icatibant use were associated with a significantly reduced rate of reported COVID-19. The results implicated potential roles for the complement cascade and tissue kallikrein-kinin pathways in the pathogenesis of COVID-19 in patients with HAE-C1INH.
Collapse
Affiliation(s)
- Camila Lopes Veronez
- From the Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, California; and
| | - Sandra C. Christiansen
- From the Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, California; and
| | - Tukisa D. Smith
- From the Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, California; and
| | - Marc A. Riedl
- From the Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, California; and
| | - Bruce L. Zuraw
- From the Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, California; and
| |
Collapse
|
11
|
Hernández-Huerta MT, Pérez-Santiago AD, Pérez-Campos Mayoral L, Sánchez Navarro LM, Rodal Canales FJ, Majluf-Cruz A, Matias-Cervantes CA, Pérez-Campos Mayoral E, Romero Díaz C, Mayoral-Andrade G, Martínez Cruz M, Luna Ángel J, Pérez-Campos E. Mechanisms of Immunothrombosis by SARS-CoV-2. Biomolecules 2021; 11:1550. [PMID: 34827548 PMCID: PMC8615366 DOI: 10.3390/biom11111550] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 12/20/2022] Open
Abstract
SARS-CoV-2 contains certain molecules that are related to the presence of immunothrombosis. Here, we review the pathogen and damage-associated molecular patterns. We also study the imbalance of different molecules participating in immunothrombosis, such as tissue factor, factors of the contact system, histones, and the role of cells, such as endothelial cells, platelets, and neutrophil extracellular traps. Regarding the pathogenetic mechanism, we discuss clinical trials, case-control studies, comparative and translational studies, and observational studies of regulatory or inhibitory molecules, more specifically, extracellular DNA and RNA, histones, sensors for RNA and DNA, as well as heparin and heparinoids. Overall, it appears that a network of cells and molecules identified in this axis is simultaneously but differentially affecting patients at different stages of COVID-19, and this is characterized by endothelial damage, microthrombosis, and inflammation.
Collapse
Affiliation(s)
- María Teresa Hernández-Huerta
- CONACyT, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.T.H.-H.); (C.A.M.-C.)
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
| | | | - Laura Pérez-Campos Mayoral
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico;
| | | | - Francisco Javier Rodal Canales
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico;
| | | | - Carlos Alberto Matias-Cervantes
- CONACyT, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.T.H.-H.); (C.A.M.-C.)
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
| | - Eduardo Pérez-Campos Mayoral
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico;
| | - Carlos Romero Díaz
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico;
| | - Gabriel Mayoral-Andrade
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico;
| | - Margarito Martínez Cruz
- Tecnológico Nacional de México/IT Oaxaca, Oaxaca de Juárez, Oaxaca 68030, Mexico; (A.D.P.-S.); (M.M.C.)
| | - Judith Luna Ángel
- Hospital General Dr. Aurelio Valdivieso, Oaxaca de Juárez, Oaxaca 68000, Mexico;
| | - Eduardo Pérez-Campos
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
- Tecnológico Nacional de México/IT Oaxaca, Oaxaca de Juárez, Oaxaca 68030, Mexico; (A.D.P.-S.); (M.M.C.)
- Laboratorio de Patología Clinica “Eduardo Pérez Ortega”, Oaxaca de Juárez, Oaxaca 68000, Mexico
| |
Collapse
|
12
|
Lignosulfonic Acid Sodium Is a Noncompetitive Inhibitor of Human Factor XIa. Pharmaceuticals (Basel) 2021; 14:ph14090886. [PMID: 34577586 PMCID: PMC8466798 DOI: 10.3390/ph14090886] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/28/2021] [Accepted: 08/29/2021] [Indexed: 12/20/2022] Open
Abstract
The anticoagulant activity of lignosulfonic acid sodium (LSAS), a non-saccharide heparin mimetic, was investigated in this study. LSAS is a relatively safe industrial byproduct with similar polyanionic characteristics to that of heparin. Human plasma clotting assays, fibrin polymerization testing, and enzyme inhibition assays were exploited to investigate the anticoagulant activity of LSAS. In normal human plasma, LSAS selectively doubled the activated partial thromboplastin time (APTT) at ~308 µg/mL. Equally, LSAS doubled APTT at ~275 µg/mL in antithrombin-deficient plasma. Yet, LSAS doubled APTT at a higher concentration of 429 µg/mL using factor XI-deficient plasma. LSAS did not affect FXIIIa-mediated fibrin polymerization at 1000 µg/mL. Enzyme assays revealed that LSAS inhibits factor XIa (FXIa) with an IC50 value of ~8 μg/mL. LSAS did not inhibit thrombin, factor IXa, factor Xa, factor XIIIa, chymotrypsin, or trypsin at the highest concentrations tested and demonstrated significant selectivity against factor XIIa and plasmin. In Michaelis–Menten kinetics, LSAS decreased the VMAX of FXIa hydrolysis of a tripeptide chromogenic substrate without significantly changing its KM indicating an allosteric inhibition mechanism. The inhibitor also disrupted the generation of FXIa–antithrombin complex, inhibited factor XIIa-mediated and thrombin-mediated activation of the zymogen factor XI to FXIa, and competed with heparin for binding to FXIa. Its action appears to be reversed by protamine sulfate. Structure–activity relationship studies demonstrated the advantageous selectivity and allosteric behavior of LSAS over the acetylated and desulfonated derivatives of LSAS. LSAS is a sulfonated heparin mimetic that demonstrates significant anticoagulant activity in human plasma. Overall, it appears that LSAS is a potent, selective, and allosteric inhibitor of FXIa with significant anticoagulant activity in human plasma. Altogether, this study introduces LSAS as a promising lead for further development as an anticoagulant.
Collapse
|
13
|
Fernandes M, Brábek J. COVID-19, corticosteroids and public health: a reappraisal. Public Health 2021; 197:48-55. [PMID: 34325124 PMCID: PMC8180552 DOI: 10.1016/j.puhe.2021.05.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 05/04/2021] [Accepted: 05/25/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To assess whether regulatory guidance on the use of dexamethasone in hospitalised COVID-19 patients is applicable to the larger population of COVID-19 cases. The surge in worldwide demand for dexamethasone suggests that the guidance, although correct, has not emphasised the danger of its wider use. STUDY DESIGN Data from the Randomised Evaluation of COVID-19 Therapy (RECOVERY) trial and the World Health Organisation (WHO) prospective meta-analysis have been deconstructed and analysed. METHODS To provide context, relevant publications were identified in PubMed using the following keywords: COVID-19, RECOVERY trial, WHO meta-analysis, variants, immunity, public health. RESULTS The WHO guidance 'Corticosteroids for COVID-19' was based on their prospective meta-analysis. This meta-analysis was weighted by data from the RECOVERY trial. CONCLUSIONS In terms of COVID-19, dexamethasone has value in a narrow indication, namely, in hospitalised patients requiring respiratory support. The media blitz likely resulted in the wider use of dexamethasone in outpatients and as a preventive medication. This is reflected in the surge in worldwide demand for dexamethasone. We ask whether the use of steroids, beyond regulatory indications, may be responsible for the recent increase in mortality and especially the emergence of mucormycosis? From the public health standpoint, the current guidance for use of dexamethasone in COVID-19 could benefit from clarification and the addition of a cautionary note.
Collapse
Affiliation(s)
- M Fernandes
- Medbase, 114 Milton Avenue, Chapel Hill, NC, 27514, USA.
| | - J Brábek
- Department of Cell Biology, Charles University, Viničná 7, Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, Vestec U Prahy, 25242, Czech Republic
| |
Collapse
|
14
|
Contact Activation Inhibitor, AB023, in Heparin-Free Hemodialysis: Results of a Randomized Phase 2 Clinical Trial. Blood 2021; 138:2173-2184. [PMID: 34086880 DOI: 10.1182/blood.2021011725] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/24/2021] [Indexed: 11/20/2022] Open
Abstract
End-stage renal disease (ESRD) patients on chronic hemodialysis have repeated blood exposure to artificial surfaces that can trigger clot formation within the hemodialysis circuit. Dialyzer clotting can lead to anemia despite erythropoietin and iron supplementation. Unfractionated heparin prevents clotting during hemodialysis, but it is not tolerated by all patients. Although heparin-free dialysis is performed, intradialytic blood entrapment can be problematic. To address this issue, we performed a randomized, double-blind, phase 2 study comparing AB023, a unique antibody that binds factor (F) XI and blocks its activation by factor XIIa but not by thrombin, to placebo in 24 patients with ESRD undergoing heparin-free hemodialysis (www.clinicaltrials.gov #NCT03612856). Patients were randomized to receive a single pre-dialysis dose of AB023 (0.25 or 0.5 mg/kg) or placebo in a 2:1 ratio and safety and preliminary efficacy were compared to placebo and to observations made prior to dosing within each treatment arm. AB023 administration was not associated with impaired hemostasis or other drug-related adverse events. Occlusive events requiring hemodialysis circuit exchange were less frequent and levels of thrombin-antithrombin complexes and C-reactive protein were lower after AB023 administration compared with data collected prior to dosing. AB023 also reduced potassium and iron entrapment in the dialyzers, consistent with less blood accumulation within the dialyzers. We conclude that despite the small sample size, inhibition of contact activation-induced coagulation with AB023 was well tolerated and reduced clotting within the dialyzer.
Collapse
|
15
|
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
|
16
|
Manzoor D, Bui C, Makhoul E, Luthringer D, Marchevsky A, Volod O. Improvement in plasma D-dimer level in severe SARS-CoV-2 infection can be an indicator of fibrinolysis suppression: Case reports. Medicine (Baltimore) 2021; 100:e25255. [PMID: 33847623 PMCID: PMC8052008 DOI: 10.1097/md.0000000000025255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/22/2021] [Accepted: 03/04/2021] [Indexed: 01/16/2023] Open
Abstract
RATIONALE Fibrinolysis shutdown associated with severe thrombotic complications is a recently recognized syndrome that was previously seldom investigated in patients with severe severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. It presents a unique therapeutic dilemma, as anticoagulation with heparin alone is insufficient to address the imbalance in fibrinolysis. And while the use of fibrinolytic agents could limit the disease severity, it is often associated with bleeding complications. There is a need for biomarkers that will guide the timely stratification of patients into those who may benefit from both anticoagulant and fibrinolytic therapies. PATIENT CONCERNS All 3 patients presented with shortness of breath along with comorbidities predisposing them to severe SARS-CoV-2 infection. One patient (Patient 3) also suffered from bilateral deep venous thrombosis. DIAGNOSES All 3 patients tested positive for SARS-CoV-2 RNA by reverse transcription polymerase chain reaction (RT-PCR) and were eventually diagnosed with respiratory failure necessitating intubation. INTERVENTIONS All 3 patients required mechanical ventilation support, 2 of which also required renal replacement therapy. All 3 patients were also placed on anticoagulation therapy. OUTCOMES In Patients 1 and 2, the initial D-dimer levels of 0.97 μg/ml fibrinogen equivalent units (FEU) and 0.83 μg/ml FEU were only slightly elevated (normal <0.50 μg/ml FEU). They developed rising D-dimer levels to a peak of 13.21 μg/ml FEU and >20.0 μg/ml FEU, respectively, which dropped to 1.34 μg/ml FEU 8 days later in Patient 1 and to 2.94 μg/ml on hospital day 13 in Patient 2. In Patient 3, the D-dimer level on admission was found to be elevated to >20.00 μg/ml FEU together with imaging evidence of thrombosis. And although he received therapeutic heparin infusion, he still developed pulmonary embolism (PE) and his D-dimer level declined to 5.91 μg/ml FEU. Despite "improvement" in their D-dimer levels, all 3 patients succumbed to multi-system organ failure. On postmortem examination, numerous arterial and venous thromboses of varying ages, many consisting primarily of fibrin, were identified in the lungs of all patients. LESSONS High D-dimer levels, with subsequent downtrend correlating with clinical deterioration, seems to be an indicator of fibrinolysis suppression. These findings can help form a hypothesis, as larger cohorts are necessary to demonstrate their reproducibility.
Collapse
|
17
|
Dhillon A, Deme JC, Furlong E, Roem D, Jongerius I, Johnson S, Lea SM. Molecular Basis for Bordetella pertussis Interference with Complement, Coagulation, Fibrinolytic, and Contact Activation Systems: the Cryo-EM Structure of the Vag8-C1 Inhibitor Complex. mBio 2021; 12:e02823-20. [PMID: 33758081 PMCID: PMC8092270 DOI: 10.1128/mbio.02823-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 02/18/2021] [Indexed: 12/27/2022] Open
Abstract
Complement, contact activation, coagulation, and fibrinolysis are serum protein cascades that need strict regulation to maintain human health. Serum glycoprotein, a C1 inhibitor (C1-INH), is a key regulator (inhibitor) of serine proteases of all the above-mentioned pathways. Recently, an autotransporter protein, virulence-associated gene 8 (Vag8), produced by the whooping cough pathogen, Bordetella pertussis, was shown to bind to C1-INH and interfere with its function. Here, we present the structure of the Vag8-C1-INH complex determined using cryo-electron microscopy at a 3.6-Å resolution. The structure shows a unique mechanism of C1-INH inhibition not employed by other pathogens, where Vag8 sequesters the reactive center loop of C1-INH, preventing its interaction with the target proteases.IMPORTANCE The structure of a 10-kDa protein complex is one of the smallest to be determined using cryo-electron microscopy at high resolution. The structure reveals that C1-INH is sequestered in an inactivated state by burial of the reactive center loop in Vag8. By so doing, the bacterium is able to simultaneously perturb the many pathways regulated by C1-INH. Virulence mechanisms such as the one described here assume more importance given the emerging evidence about dysregulation of contact activation, coagulation, and fibrinolysis leading to COVID-19 pneumonia.
Collapse
Affiliation(s)
- Arun Dhillon
- Sir William Dunn School of Pathology, Oxford, United Kingdom
| | - Justin C Deme
- Sir William Dunn School of Pathology, Oxford, United Kingdom
- Central Oxford Structural Molecular Imaging Centre, Oxford, United Kingdom
| | - Emily Furlong
- Sir William Dunn School of Pathology, Oxford, United Kingdom
| | - Dorina Roem
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | - Ilse Jongerius
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
- Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Steven Johnson
- Sir William Dunn School of Pathology, Oxford, United Kingdom
| | - Susan M Lea
- Sir William Dunn School of Pathology, Oxford, United Kingdom
- Central Oxford Structural Molecular Imaging Centre, Oxford, United Kingdom
| |
Collapse
|
18
|
Lipcsey M, Persson B, Eriksson O, Blom AM, Fromell K, Hultström M, Huber-Lang M, Ekdahl KN, Frithiof R, Nilsson B. The Outcome of Critically Ill COVID-19 Patients Is Linked to Thromboinflammation Dominated by the Kallikrein/Kinin System. Front Immunol 2021; 12:627579. [PMID: 33692801 PMCID: PMC7937878 DOI: 10.3389/fimmu.2021.627579] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/18/2021] [Indexed: 01/08/2023] Open
Abstract
An important manifestation of severe COVID-19 is the ARDS-like lung injury that is associated with vascular endothelialitis, thrombosis, and angiogenesis. The intravascular innate immune system (IIIS), including the complement, contact, coagulation, and fibrinolysis systems, which is crucial for recognizing and eliminating microorganisms and debris in the body, is likely to be involved in the pathogenesis of COVID-19 ARDS. Biomarkers for IIIS activation were studied in the first 66 patients with COVID-19 admitted to the ICU in Uppsala University Hospital, both cross-sectionally on day 1 and in 19 patients longitudinally for up to a month, in a prospective study. IIIS analyses were compared with biochemical parameters and clinical outcome and survival. Blood cascade systems activation leading to an overreactive conjunct thromboinflammation was demonstrated, reflected in consumption of individual cascade system components, e.g., FXII, prekallikrein, and high molecular weight kininogen and in increased levels of activation products, e.g., C4d, C3a, C3d,g, sC5b-9, TAT, and D-dimer. Strong associations were found between the blood cascade systems and organ damage, illness severity scores, and survival. We show that critically ill COVID-19 patients display a conjunct activation of the IIIS that is linked to organ damage of the lung, heart, kidneys, and death. We present evidence that the complement and in particular the kallikrein/kinin system is strongly activated and that both systems are prognostic markers of the outcome of the patients suggesting their role in driving the inflammation. Already licensed kallikrein/kinin inhibitors are potential drugs for treatment of critically ill patients with COVID-19.
Collapse
Affiliation(s)
- Miklós Lipcsey
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
- Hedenstierna Laboratory, Anesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Barbro Persson
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Oskar Eriksson
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Anna M. Blom
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Karin Fromell
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Michael Hultström
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
- Unit for Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Kristina N. Ekdahl
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Robert Frithiof
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Bo Nilsson
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
19
|
A novel rationale for targeting FXI: Insights from the hemostatic microRNA targetome for emerging anticoagulant strategies. Pharmacol Ther 2021; 218:107676. [DOI: 10.1016/j.pharmthera.2020.107676] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
|
20
|
Kar S, Mottamal M, Al‐Horani RA. Discovery of Benzyl Tetraphosphonate Derivative as Inhibitor of Human Factor Xia. ChemistryOpen 2020; 9:1161-1172. [PMID: 33204588 PMCID: PMC7654249 DOI: 10.1002/open.202000277] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/08/2020] [Indexed: 12/14/2022] Open
Abstract
The inhibition of factor XIa (FXIa) is a trending paradigm for the development of new generations of anticoagulants without a substantial risk of bleeding. In this report, we present the discovery of a benzyl tetra-phosphonate derivative as a potent and selective inhibitor of human FXIa. Biochemical screening of four phosphonate/phosphate derivatives has led to the identification of the molecule that inhibited human FXIa with an IC50 value of ∼7.4 μM and a submaximal efficacy of ∼68 %. The inhibitor was at least 14-fold more selective to FXIa over thrombin, factor IXa, factor Xa, and factor XIIIa. It also inhibited FXIa-mediated activation of factor IX and prolonged the activated partial thromboplastin time of human plasma. In Michaelis-Menten kinetics experiment, inhibitor 1 reduced the VMAX of FXIa hydrolysis of a chromogenic substrate without significantly affecting its KM suggesting an allosteric mechanism of inhibition. The inhibitor also disrupted the formation of FXIa - antithrombin complex and inhibited thrombin-mediated and factor XIIa-mediated formation of FXIa from its zymogen factor XI. Inhibitor 1 has been proposed to bind to or near the heparin/polyphosphate-binding site in the catalytic domain of FXIa. Overall, inhibitor 1 is the first benzyl tetraphosphonate small molecule that allosterically inhibits human FXIa, blocks its physiological function, and prevents its zymogen activation by other clotting factors under in vitro conditions. Thus, we put forward benzyl tetra-phosphonate 1 as a novel lead inhibitor of human FXIa to guide future efforts in the development of allosteric anticoagulants.
Collapse
Affiliation(s)
- Srabani Kar
- Division of Basic Pharmaceutical Sciences College of PharmacyXavier University of LouisianaNew OrleansLA70125USA
| | - Madhusoodanan Mottamal
- RCMI Cancer Research Center & Department of ChemistryXavier University of LouisianaNew OrleansLA70125USA
| | - Rami A. Al‐Horani
- Division of Basic Pharmaceutical Sciences College of PharmacyXavier University of LouisianaNew OrleansLA70125USA
| |
Collapse
|
21
|
Frydman GH, Streiff MB, Connors JM, Piazza G. The Potential Role of Coagulation Factor Xa in the Pathophysiology of COVID-19: A Role for Anticoagulants as Multimodal Therapeutic Agents. ACTA ACUST UNITED AC 2020; 4:e288-e299. [PMID: 33043235 PMCID: PMC7541169 DOI: 10.1055/s-0040-1718415] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023]
Abstract
SARS-CoV-2 infection (COVID-19) results in local and systemic activation of inflammation and coagulation. In this review article, we will discuss the potential role of coagulation factor Xa (FXa) in the pathophysiology of COVID-19. FXa, a serine protease, has been shown to play a role in the cleavage of SARS-CoV-1 spike protein (SP), with the inhibition of FXa resulting in the inhibition of viral infectivity. FX is known to be primarily produced in the liver, but it is also expressed by multiple cells types, including alveolar epithelium, cardiac myocytes, and macrophages. Considering that patients with preexisting conditions, including cardiopulmonary disease, are at an increased risk of severe COVID-19, we discuss the potential role of increased levels of FX in these patients, resulting in a potential increased propensity to have a higher infectious rate and viral load, increased activation of coagulation and inflammation, and development of fibrosis. With these observations in mind, we postulate as to the potential therapeutic role of FXa inhibitors as a prophylactic and therapeutic treatment for high-risk patients with COVID-19.
Collapse
Affiliation(s)
- Galit H Frydman
- Coagulo Medical Technologies, Inc., Auburndale, Massachusetts, United States.,Center for Biomedical Engineering, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States.,Division of Trauma, Emergency Surgery and Surgical Critical Care, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Michael B Streiff
- Division of Hematology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Jean M Connors
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Gregory Piazza
- Division of Cardiovascular Medicine Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States
| |
Collapse
|
22
|
Parra-Izquierdo I, Aslan JE. Perspectives on Platelet Heterogeneity and Host Immune Response in Coronavirus Disease 2019 (COVID-19). Semin Thromb Hemost 2020; 46:826-830. [PMID: 32882716 PMCID: PMC7645832 DOI: 10.1055/s-0040-1715093] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Iván Parra-Izquierdo
- Knight Cardiovascular Institute, Division of Cardiology, Department of Medicine, School of Medicine, Oregon Health and Science University, Portland, Oregon
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Joseph E. Aslan
- Knight Cardiovascular Institute, Division of Cardiology, Department of Medicine, School of Medicine, Oregon Health and Science University, Portland, Oregon
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, Oregon
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
23
|
Al-Benna S. Pathophysiology of coronavirus disease 2019 for wound care professionals. Int Wound J 2020; 17:1935-1940. [PMID: 32986928 PMCID: PMC7536990 DOI: 10.1111/iwj.13483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/30/2020] [Indexed: 12/13/2022] Open
Abstract
There is pressing urgency to understand the pathogenesis of the severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) which causes Coronavirus disease 2019 (COVID‐19). The tissue tropism of SARS‐CoV‐2 includes not only the lung but also the vascular and integumentary systems. Angiotensin‐converting enzyme 2 (ACE2) appears to be the key functional receptor for the virus. There is a prominent innate immune response to SARS‐CoV‐2 infection, including inflammatory cytokines, chemokines, the complement system, and acute phase proteins. The pathophysiologic significance of SARS‐COV‐2 and host immune system interaction, and COVID‐19‐associated coagulopathy instigating microvascular injury syndrome mediated by activation of complement pathways, and an associated procoagulant state is important for wound care professionals to understand.
Collapse
Affiliation(s)
- Sammy Al-Benna
- Division of Plastic and Reconstructive Surgery, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Academic Hospital, Cape Town, South Africa
| |
Collapse
|
24
|
Andreani G, Uscello L, Montaruli B, Briozzo A, Vitale F, Tricarico M, Arnaldi L, Marengo S, Norbiato C. Acquired Factor XI Deficiency during SARS-CoV-2 Infection: Not Only Thrombosis. TH OPEN 2020; 4:e233-e235. [PMID: 32939441 PMCID: PMC7487214 DOI: 10.1055/s-0040-1714696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/25/2020] [Indexed: 01/01/2023] Open
Affiliation(s)
- Giacomo Andreani
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Lorenzo Uscello
- Department of Internal Medicine, A.O. Ordine Mauriziano, Turin, Italy
| | | | - Antonio Briozzo
- Department of Internal Medicine, A.O. Ordine Mauriziano, Turin, Italy
| | - Francesco Vitale
- Department of Internal Medicine, A.O. Ordine Mauriziano, Turin, Italy
| | | | - Luisa Arnaldi
- Department of Internal Medicine, A.O. Ordine Mauriziano, Turin, Italy
| | - Stefania Marengo
- Department of Internal Medicine, A.O. Ordine Mauriziano, Turin, Italy
| | - Claudio Norbiato
- Department of Internal Medicine, A.O. Ordine Mauriziano, Turin, Italy
| |
Collapse
|
25
|
Xu Y, Liu S, Zhang Y, Zhi Y. Does hereditary angioedema make COVID-19 worse? World Allergy Organ J 2020; 13:100454. [PMID: 32834893 PMCID: PMC7416729 DOI: 10.1016/j.waojou.2020.100454] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 07/31/2020] [Indexed: 12/19/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has spread rapidly worldwide. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent for COVID-19, enters host cells via angiotensin-converting enzyme 2 (ACE2) and depletes ACE2, which is necessary for bradykinin metabolism. The depletion of ACE2 results in the accumulation of des-Arg (9)-bradykinin and possible bradykinin, both of which bind to bradykinin receptors and induce vasodilation, lung injury, and inflammation. It is well known that an overactivated contact system and excessive production of bradykinin comprise the key mechanisms that drive the pathogenesis of hereditary angioedema (HAE). It is reasonable to speculate that COVID-19 may increase disease activity in patients with HAE and vice versa. In this review, we explore the potential interactions between COVID-19 and HAE in terms of the contact system, the complement system, cytokine release, increased T helper 17 cells, and hematologic abnormalities. We conclude with the hypothesis that comorbidity with HAE might favor COVID-19 progression and may worsen its outcomes, while COVID-19 might in turn aggravate pre-existing HAE and prompt the onset of HAE in asymptomatic carriers of HAE-related mutations. Based on the pathophysiologic links, we suggest that long-term prophylaxis should be considered in patients with HAE at risk of SARS-CoV-2 infection, especially the prophylactic use of C1 inhibitor and lanadelumab and that HAE patients must have medications for acute attacks of angioedema. Additionally, therapeutic strategies employed in HAE should be considered for the treatment of COVID-19, and clinical trials should be performed.
Collapse
Key Words
- ACE2
- ADAM metallopeptidase domain 17, ADAM17
- C1 inhibitor, C1–INH
- COVID-19
- Complement system
- Contact system
- Coronavirus disease 2019, COVID-19
- Hereditary angioedema
- Middle East respiratory syndrome coronavirus, MERS-CoV
- acute respiratory distress syndrome, ARDS
- angiotensin-converting enzyme, ACE
- bradykinin receptor B1, B1R
- bradykinin receptor B2, B2R
- bradykinin, BK
- des-Arg(9)-bradykinin, DABK
- granulocyte-colony stimulating factor, GCSF
- granulocyte-macrophage colony stimulating factor, GM-CSF
- hereditary angioedema, HAE
- high-molecular-weight kininogen, HMWK
- interleukin-1, IL-1
- macrophage inflammatory protein, MIP
- mannose-binding lectin associated serine protease, MASP
- mannose-binding lectin, MBL
- severe acute respiratory syndrome coronavirus 2, SARS-CoV-2
- transforming growth factor-β, TGF-β
- transmembrane serine protease, TMPRSS2
- tumor necrosis factor γ, TNF-γ
Collapse
Affiliation(s)
- Yingyang Xu
- Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.,Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment on Allergic Diseases, Beijing, 100730, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, 100730, China.,Centre for Translational Medicine, Peking Union Medical College Hospital, China
| | - Shuang Liu
- School of Clinical Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Yan Zhang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Yuxiang Zhi
- Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.,Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment on Allergic Diseases, Beijing, 100730, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, 100730, China.,Centre for Translational Medicine, Peking Union Medical College Hospital, China
| |
Collapse
|
26
|
Lakshmanan HHS, Pore AA, Kohs TCL, Yazar F, Thompson RM, Jurney PL, Maddala J, Olson SR, Shatzel JJ, Vanapalli SA, McCarty OJT. Design of a Microfluidic Bleeding Chip to Evaluate Antithrombotic Agents for Use in COVID-19 Patients. Cell Mol Bioeng 2020; 13:331-339. [PMID: 32837586 PMCID: PMC7408976 DOI: 10.1007/s12195-020-00644-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/31/2020] [Indexed: 12/27/2022] Open
Abstract
Introduction Interventions that could prevent thrombosis, clinical decompensation, and respiratory compromise in patients with novel coronavirus disease (COVID-19) are key to decrease mortality rate. Studies show that profound cytokine release and excessive activation of blood coagulation appear to be key drivers of COVID-19 associated mortality. Since limited in vitro methods exist for assessing the effects of anticoagulants on hemostasis, the development of novel therapies to safely prevent thrombosis in COVID-19 patients relies on preclinical animal models and early phase human trials. Herein we present the design of a microfluidic “bleeding chip” to evaluate the effects of antithrombotic therapies on hemostatic plug formation in vitro. Methods The design of the microfluidic device consists of two orthogonal channels: an inlet that serves as a model blood vessel, and a bleeding channel to model hemostatic plug formation at sites of compromised endothelial barrier function. This is achieved by placing a series of 3 pillars spaced 10 μm apart at the intersection of the two channels. The pillars and bleeding channel are coated with the extracellular matrix protein collagen. Results Perfusion of human whole blood through the microfluidic bleeding chip led to initial platelet adhesion and aggregation at the pillars followed by hemostatic plug formation and occlusion of the bleeding channel. Conclusions Safe and effective mitigating agents are needed for treatment and prevention of thrombotic complications in COVID-19 patients. This simple microfluidic device holds potential to be developed into a tool for assessing the effects of anticoagulant therapy on hemostasis.
Collapse
Affiliation(s)
- Hari Hara Sudhan Lakshmanan
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA
| | - Adity A Pore
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX USA
| | - Tia C L Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA
| | - Feyza Yazar
- Department of Biomedical Engineering, San José State University, San Jose, CA USA
| | - Rachel M Thompson
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA
| | - Patrick L Jurney
- Department of Biomedical Engineering, San José State University, San Jose, CA USA
| | - Jeevan Maddala
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA
| | - Sven R Olson
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA.,Division of Hematology & Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR USA
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA.,Division of Hematology & Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR USA
| | - Siva A Vanapalli
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave; CH13B, Portland, OR 97239 USA.,Division of Hematology & Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR USA
| |
Collapse
|
27
|
Kusadasi N, Sikma M, Huisman A, Westerink J, Maas C, Schutgens R. A Pathophysiological Perspective on the SARS-CoV-2 Coagulopathy. Hemasphere 2020; 4:e457. [PMID: 32885147 PMCID: PMC7430228 DOI: 10.1097/hs9.0000000000000457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/22/2020] [Indexed: 01/08/2023] Open
Abstract
Recent evidence is focusing on the presence of a hypercoagulable state with development of both venous and arterial thromboembolic complications in patients infected with SARS-CoV-2. The ongoing activation of coagulation related to the severity of the illness is further characterized by thrombotic microangiopathy and endotheliitis. These microangiopathic changes cannot be classified as classical disseminated intravascular coagulation (DIC). In this short review we describe the interaction between coagulation and inflammation with focus on the possible mechanisms that might be involved in SARS-CoV-2 infection associated coagulopathy in the critically ill.
Collapse
Affiliation(s)
- Nuray Kusadasi
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maaike Sikma
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Dutch Poisons Information Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Albert Huisman
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan Westerink
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Coen Maas
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roger Schutgens
- Van Creveldkliniek, Benign Hematology Center, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
28
|
Ribes A, Vardon-Bounes F, Mémier V, Poette M, Au-Duong J, Garcia C, Minville V, Sié P, Bura-Rivière A, Voisin S, Payrastre B. Thromboembolic events and Covid-19. Adv Biol Regul 2020; 77:100735. [PMID: 32773098 PMCID: PMC7833411 DOI: 10.1016/j.jbior.2020.100735] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 01/08/2023]
Abstract
The novel Corona virus infection (Covid-19) first identified in China in December 2019 has rapidly progressed in pandemic leading to significant mortality and unprecedented challenge for healthcare systems. Although the clinical spectrum of Covid-19 is variable, acute respiratory failure and systemic coagulopathy are common in severe Covid-19 patients. Lung is an important target of the SARS-CoV-2 virus causing eventually acute respiratory distress syndrome associated to a thromboinflammatory state. The cytokinic storm, thromboinflammation and pulmonary tropism are the bedrock of tissue lesions responsible for acute respiratory failure and for prolonged infection that may lead to multiple organ failure and death. The thrombogenicity of this infectious disease is illustrated by the high frequency of thromboembolic events observed even in Covid-19 patients treated with anticoagulation. Increased D-Dimers, a biomarker reflecting activation of hemostasis and fibrinolysis, and low platelet count (thrombocytopenia) are associated with higher mortality in Covid-19 patients. In this review, we will summarize our current knowledge on the thromboembolic manifestations, the disturbed hemostatic parameters, and the thromboinflammatory conditions associated to Covid-19 and we will discuss the modalities of anticoagulant treatment or other potential antithrombotic options.
Collapse
Affiliation(s)
- Agnès Ribes
- Inserm U1048 and Université Toulouse III Paul Sabatier, I2MC, 31024, Toulouse Cedex 03, France; Laboratoire d'Hématologie, CHU de Toulouse, 31059, Toulouse, France
| | - Fanny Vardon-Bounes
- Inserm U1048 and Université Toulouse III Paul Sabatier, I2MC, 31024, Toulouse Cedex 03, France; Pôle Anesthésie-Réanimation, CHU de Toulouse, 31059, Toulouse, France
| | - Vincent Mémier
- Laboratoire d'Hématologie, CHU de Toulouse, 31059, Toulouse, France
| | - Michael Poette
- Inserm U1048 and Université Toulouse III Paul Sabatier, I2MC, 31024, Toulouse Cedex 03, France; Pôle Anesthésie-Réanimation, CHU de Toulouse, 31059, Toulouse, France
| | - Jonathan Au-Duong
- Inserm U1048 and Université Toulouse III Paul Sabatier, I2MC, 31024, Toulouse Cedex 03, France; Pôle Anesthésie-Réanimation, CHU de Toulouse, 31059, Toulouse, France
| | - Cédric Garcia
- Inserm U1048 and Université Toulouse III Paul Sabatier, I2MC, 31024, Toulouse Cedex 03, France; Laboratoire d'Hématologie, CHU de Toulouse, 31059, Toulouse, France
| | - Vincent Minville
- Pôle Anesthésie-Réanimation, CHU de Toulouse, 31059, Toulouse, France
| | - Pierre Sié
- Inserm U1048 and Université Toulouse III Paul Sabatier, I2MC, 31024, Toulouse Cedex 03, France; Laboratoire d'Hématologie, CHU de Toulouse, 31059, Toulouse, France
| | | | - Sophie Voisin
- Inserm U1048 and Université Toulouse III Paul Sabatier, I2MC, 31024, Toulouse Cedex 03, France; Laboratoire d'Hématologie, CHU de Toulouse, 31059, Toulouse, France
| | - Bernard Payrastre
- Inserm U1048 and Université Toulouse III Paul Sabatier, I2MC, 31024, Toulouse Cedex 03, France; Laboratoire d'Hématologie, CHU de Toulouse, 31059, Toulouse, France.
| |
Collapse
|
29
|
Abstract
The recent emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the ensuing global pandemic has presented a health emergency of unprecedented magnitude. Recent clinical data has highlighted that coronavirus disease 2019 (COVID-19) is associated with a significant risk of thrombotic complications ranging from microvascular thrombosis, venous thromboembolic disease, and stroke. Importantly, thrombotic complications are markers of severe COVID-19 and are associated with multiorgan failure and increased mortality. The evidence to date supports the concept that the thrombotic manifestations of severe COVID-19 are due to the ability of SARS-CoV-2 to invade endothelial cells via ACE-2 (angiotensin-converting enzyme 2), which is expressed on the endothelial cell surface. However, in patients with COVID-19 the subsequent endothelial inflammation, complement activation, thrombin generation, platelet, and leukocyte recruitment, and the initiation of innate and adaptive immune responses culminate in immunothrombosis, ultimately causing (micro)thrombotic complications, such as deep vein thrombosis, pulmonary embolism, and stroke. Accordingly, the activation of coagulation (eg, as measured with plasma D-dimer) and thrombocytopenia have emerged as prognostic markers in COVID-19. Given thrombotic complications are central determinants of the high mortality rate in COVID-19, strategies to prevent thrombosis are of critical importance. Several antithrombotic drugs have been proposed as potential therapies to prevent COVID-19-associated thrombosis, including heparin, FXII inhibitors, fibrinolytic drugs, nafamostat, and dipyridamole, many of which also possess pleiotropic anti-inflammatory or antiviral effects. The growing awareness and mechanistic understanding of the prothrombotic state of COVID-19 patients are driving efforts to more stringent diagnostic screening for thrombotic complications and to the early institution of antithrombotic drugs, for both the prevention and therapy of thrombotic complications. The shifting paradigm of diagnostic and treatment strategies holds significant promise to reduce the burden of thrombotic complications and ultimately improve the prognosis for patients with COVID-19.
Collapse
Affiliation(s)
- James D. McFadyen
- From the Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
- Clinical Hematology Department (J.D.M., H.S.), Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
| | - Hannah Stevens
- From the Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
- Clinical Hematology Department (J.D.M., H.S.), Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
| | - Karlheinz Peter
- Department of Cardiology (K.P.), Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
| |
Collapse
|
30
|
Thachil J, Cushman M, Srivastava A. A proposal for staging COVID-19 coagulopathy. Res Pract Thromb Haemost 2020; 4:731-736. [PMID: 32685880 PMCID: PMC7272892 DOI: 10.1002/rth2.12372] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/05/2020] [Accepted: 05/08/2020] [Indexed: 12/19/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is associated with significant hypercoagulability. However, despite prophylactic anticoagulation, critically ill patients with this condition develop thromboses. This forum discusses the lungs as the epicenter for the hemostatic issues, puts forward a proposal for staging COVID-19 coagulopathy based on available diagnostic markers, and suggest considering current and future treatment options based on these different stages.
Collapse
Affiliation(s)
- Jecko Thachil
- Department of HaematologyManchester University HospitalsManchesterUK
| | - Mary Cushman
- Department of MedicineLarner College of MedicineUniversity of VermontBurlingtonVTUSA
| | - Alok Srivastava
- Department of HaematologyChristian Medical CollegeVelloreIndia
| |
Collapse
|
31
|
Bikdeli B, Madhavan MV, Gupta A, Jimenez D, Burton JR, Der Nigoghossian C, Chuich T, Nouri SN, Dreyfus I, Driggin E, Sethi S, Sehgal K, Chatterjee S, Ageno W, Madjid M, Guo Y, Tang LV, Hu Y, Bertoletti L, Giri J, Cushman M, Quéré I, Dimakakos EP, Gibson CM, Lippi G, Favaloro EJ, Fareed J, Tafur AJ, Francese DP, Batra J, Falanga A, Clerkin KJ, Uriel N, Kirtane A, McLintock C, Hunt BJ, Spyropoulos AC, Barnes GD, Eikelboom JW, Weinberg I, Schulman S, Carrier M, Piazza G, Beckman JA, Leon MB, Stone GW, Rosenkranz S, Goldhaber SZ, Parikh SA, Monreal M, Krumholz HM, Konstantinides SV, Weitz JI, Lip GYH, The Global COVID-19 Thrombosis Collaborative Group . Pharmacological Agents Targeting Thromboinflammation in COVID-19: Review and Implications for Future Research. Thromb Haemost 2020; 120:1004-1024. [PMID: 32473596 PMCID: PMC7516364 DOI: 10.1055/s-0040-1713152] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19), currently a worldwide pandemic, is a viral illness caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The suspected contribution of thrombotic events to morbidity and mortality in COVID-19 patients has prompted a search for novel potential options for preventing COVID-19-associated thrombotic disease. In this article by the Global COVID-19 Thrombosis Collaborative Group, we describe novel dosing approaches for commonly used antithrombotic agents (especially heparin-based regimens) and the potential use of less widely used antithrombotic drugs in the absence of confirmed thrombosis. Although these therapies may have direct antithrombotic effects, other mechanisms of action, including anti-inflammatory or antiviral effects, have been postulated. Based on survey results from this group of authors, we suggest research priorities for specific agents and subgroups of patients with COVID-19. Further, we review other agents, including immunomodulators, that may have antithrombotic properties. It is our hope that the present document will encourage and stimulate future prospective studies and randomized trials to study the safety, efficacy, and optimal use of these agents for prevention or management of thrombosis in COVID-19.
Collapse
Affiliation(s)
- Behnood Bikdeli
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
- Center for Outcomes Research and Evaluation (CORE), Yale School of Medicine, New Haven, Connecticut, United States
- Clinical Trials Center, Cardiovascular Research Foundation, New York, New York, United States
| | - Mahesh V. Madhavan
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
- Clinical Trials Center, Cardiovascular Research Foundation, New York, New York, United States
| | - Aakriti Gupta
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
- Center for Outcomes Research and Evaluation (CORE), Yale School of Medicine, New Haven, Connecticut, United States
- Clinical Trials Center, Cardiovascular Research Foundation, New York, New York, United States
| | - David Jimenez
- Respiratory Department, Hospital Ramón y Cajal, Madrid, Spain
- Medicine Department, Universidad de Alcalá (IRYCIS), CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - John R. Burton
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
| | - Caroline Der Nigoghossian
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
| | - Taylor Chuich
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
| | - Shayan Nabavi Nouri
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
| | - Isaac Dreyfus
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
| | - Elissa Driggin
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
| | - Sanjum Sethi
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
| | - Kartik Sehgal
- Harvard Medical School, Boston, Massachusetts, United States
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| | - Saurav Chatterjee
- North Shore and Long Island Jewish University Hospitals, Queens, New York, United States
| | - Walter Ageno
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Mohammad Madjid
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States
| | - Yutao Guo
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
| | - Liang V. Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Laurent Bertoletti
- Department of “Médecine Vasculaire et Thérapeutique,” CIC 1408, INNOVTE, CHU de St-Etienne and INSERM UMR1059, Université Jean-Monnet, Saint-Etienne, France
| | - Jay Giri
- Cardiovascular Division, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Penn Cardiovascular Outcomes, Quality, and Evaluative Research Center, Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, United States
| | - Mary Cushman
- Larner College of Medicine, University of Vermont, Burlington, Vermont, United States
| | - Isabelle Quéré
- Department of Vascular Medicine, University of Montpellier, Montpellier CHU, InnoVTE F-CRIN Network, Montpellier, France
| | | | - C. Michael Gibson
- Harvard Medical School, Boston, Massachusetts, United States
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| | - Giuseppe Lippi
- Laboratory of Clinical Chemistry and Hematology, University Hospital of Verona, Verona, Italy
| | - Emmanuel J. Favaloro
- Laboratory Haematology, Institute of Clinical Pathology and Medical Research (ICPMR), NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia
- Sydney Centres for Thrombosis and Haemostasis, Westmead, NSW, Australia
| | - Jawed Fareed
- Loyola University Medical Center, Chicago, Illinois, United States
| | - Alfonso J. Tafur
- Pritzker School of Medicine at the University of Chicago, Chicago, Illinois, United States
- Division of Vascular Medicine, Department of Medicine, NorthShore University HealthSystem, Skokie, Illinois, United States
| | - Dominic P. Francese
- Clinical Trials Center, Cardiovascular Research Foundation, New York, New York, United States
| | - Jaya Batra
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
| | - Anna Falanga
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, University of Milan Bicocca, Bergamo, Italy
| | - Kevin J. Clerkin
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
| | - Nir Uriel
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
| | - Ajay Kirtane
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
- Clinical Trials Center, Cardiovascular Research Foundation, New York, New York, United States
| | | | | | - Alex C. Spyropoulos
- The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, United States
| | - Geoffrey D. Barnes
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan, United States
- Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, United States
| | - John W. Eikelboom
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Ido Weinberg
- Harvard Medical School, Boston, Massachusetts, United States
- Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Sam Schulman
- Department of Obstetrics and Gynecology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- McMaster University, Hamilton, Ontario, Canada
- Thrombosis & Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Marc Carrier
- The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Gregory Piazza
- Harvard Medical School, Boston, Massachusetts, United States
- Brigham and Women's Hospital, Boston, Massachusetts, United States
| | - Joshua A. Beckman
- Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Martin B. Leon
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
- Clinical Trials Center, Cardiovascular Research Foundation, New York, New York, United States
| | - Gregg W. Stone
- Clinical Trials Center, Cardiovascular Research Foundation, New York, New York, United States
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Stephan Rosenkranz
- Department of Cardiology, Cologne Cardiovascular Research Center (CCRC), Heart Center at the University of Cologne, University of Cologne, Cologne, Germany
| | - Samuel Z. Goldhaber
- Harvard Medical School, Boston, Massachusetts, United States
- Brigham and Women's Hospital, Boston, Massachusetts, United States
| | - Sahil A. Parikh
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, United States
- Clinical Trials Center, Cardiovascular Research Foundation, New York, New York, United States
| | - Manuel Monreal
- Department of Internal Medicine, Hospital Universitari Germans Trials I Pujol, Universidad Católica de Murcia, Barcelona, Spain
| | - Harlan M. Krumholz
- Center for Outcomes Research and Evaluation (CORE), Yale School of Medicine, New Haven, Connecticut, United States
- Department of Health Policy and Administration, Yale School of Public Health, New Haven, Connecticut, United States
- Section of Cardiovascular Medicine, Department of Internal Medicie, Yale School of Medicine, New Haven, Connecticut, United States
| | | | - Jeffrey I. Weitz
- McMaster University, Hamilton, Ontario, Canada
- Thrombosis & Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Gregory Y. H. Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
- Department of Clinical Medicine, Aalborg Thrombosis Research Unit, Aalborg University, Aalborg, Denmark
| | | |
Collapse
|