1
|
Ware AD, Veigulis ZP, Hoover PJ, Blumke TL, Ioannou GN, Boyko EJ, Osborne TF. Incidence and risk of post-COVID-19 thromboembolic disease and the impact of aspirin prescription; nationwide observational cohort at the US Department of Veteran Affairs. PLoS One 2024; 19:e0302612. [PMID: 39288150 PMCID: PMC11407644 DOI: 10.1371/journal.pone.0302612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/25/2024] [Indexed: 09/19/2024] Open
Abstract
INTRODUCTION COVID-19 triggers prothrombotic and proinflammatory changes, with thrombotic disease prevalent in up to 30% SARS-CoV-2 infected patients. Early work suggests that aspirin could prevent COVID-19 related thromboembolic disorders in some studies but not others. This study leverages data from the largest integrated healthcare system in the United States to better understand this association. Our objective was to evaluate the incidence and risk of COVID-19 associated acute thromboembolic disorders and the potential impact of aspirin. METHODS This retrospective, observational study utilized national electronic health record data from the Veterans Health Administration. 334,374 Veterans who tested positive for COVID-19 from March 2, 2020, to June 13, 2022, were included, 81,830 of whom had preexisting aspirin prescription prior to their COVID-19 diagnosis. Patients with and without aspirin prescriptions were matched and the odds of post-COVID acute thromboembolic disorders were assessed. RESULTS 10.1% of Veterans had a documented thromboembolic disorder within 12 months following their COVID-19 diagnosis. Those with specific comorbidities were at greatest risk. Preexisting aspirin prescription was associated with a significant decrease risk of post-COVID-19 thromboembolic disorders, including pulmonary embolism (OR [95% CI]: 0.69 [0.65, 0.74]) and deep vein thrombosis (OR [95% CI]: 0.76 [0.69, 0.83], but an increased risk of acute arterial diseases, including ischemic stroke (OR [95% CI]: 1.54 [1.46, 1.60]) and acute ischemic heart disease (1.33 [1.26, 1.39]). CONCLUSIONS Findings demonstrated that preexisting aspirin prescription prior to COVID-19 diagnosis was associated with significantly decreased risk of venous thromboembolism and pulmonary embolism but increased risk of acute arterial disease. The risk of arterial disease may be associated with increased COVID-19 prothrombotic effects superimposed on preexisting chronic cardiovascular disease for which aspirin was already prescribed. Prospective clinical trials may help to further assess the efficacy of aspirin use prior to COVID-19 diagnosis for the prevention of post-COVID-19 thromboembolic disorders.
Collapse
Affiliation(s)
- Anna D. Ware
- National Center for Collaborative Healthcare Innovation, VA Palo Alto Healthcare System, Palo Alto, California, United States of America
| | - Zachary P. Veigulis
- National Center for Collaborative Healthcare Innovation, VA Palo Alto Healthcare System, Palo Alto, California, United States of America
- Department of Business Analytics, University of Iowa Tippie College of Business, Iowa City, Iowa, United States of America
| | - Peter J. Hoover
- National Center for Collaborative Healthcare Innovation, VA Palo Alto Healthcare System, Palo Alto, California, United States of America
| | - Terri L. Blumke
- National Center for Collaborative Healthcare Innovation, VA Palo Alto Healthcare System, Palo Alto, California, United States of America
| | - George N. Ioannou
- Center of Innovation for Veteran-Centered and Value-Driven Care, Veterans Affairs Puget Sound Healthcare System, Seattle, Washington, United States of America
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Edward J. Boyko
- Seattle Epidemiologic Research and Information Center, Veterans Affairs Puget Sound Healthcare System, Seattle, Washington, United States of America
| | - Thomas F. Osborne
- National Center for Collaborative Healthcare Innovation, VA Palo Alto Healthcare System, Palo Alto, California, United States of America
- Department of Radiology, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
2
|
Liu H, Brostoff T, Ramirez A, Wong T, Rowland DJ, Heffner M, Flores A, Willis B, Evans JJ, Lanoue L, Lloyd KCK, Coffey LL. Establishment and characterization of an h ACE2/hTMPRSS2 knock-in mouse model to study SARS-CoV-2. Front Immunol 2024; 15:1428711. [PMID: 39050847 PMCID: PMC11266032 DOI: 10.3389/fimmu.2024.1428711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Despite a substantial body of research, we lack fundamental understanding of the pathophysiology of COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) including pulmonary and cardiovascular outcomes, in part due to limitations of murine models. Most models use transgenic mice (K18) that express the human (h) angiotensin converting enzyme 2 (ACE2), ACE2 knock-in (KI) mice, or mouse-adapted strains of SARS-CoV-2. Further, many SARS-CoV-2 variants produce fatal neurologic disease in K18 mice and most murine studies focus only on acute disease in the first 14 days post inoculation (dpi). To better enable understanding of both acute (<14 dpi) and post-acute (>14 dpi) infection phases, we describe the development and characterization of a novel non-lethal KI mouse that expresses both the ACE2 and transmembrane serine protease 2 (TMPRSS2) genes (hACE2/hTMPRSS2). The human genes were engineered to replace the orthologous mouse gene loci but remain under control of their respective murine promoters, resulting in expression of ACE2 and TMPRSS2 instead of their murine counterparts. After intranasal inoculation with an omicron strain of SARS-CoV-2, hACE2/hTMPRSS2 KI mice transiently lost weight but recovered by 7 dpi. Infectious SARS-CoV-2 was detected in nasopharyngeal swabs 1-2 dpi and in lung tissues 2-6 dpi, peaking 4 dpi. These outcomes were similar to those in K18 mice that were inoculated in parallel. To determine the extent to which hACE2/hTMPRSS2 KI mice are suitable to model pulmonary and cardiovascular outcomes, physiological assessments measuring locomotion, behavior and reflexes, biomonitoring to measure cardiac activity and respiration, and micro computed tomography to assess lung function were conducted frequently to 6 months post inoculation. Male but not female SARS-CoV-2 inoculated hACE2/hTMPRSS2 KI mice showed a transient reduction in locomotion compared to control saline treated mice. No significant changes in respiration, oxygen saturation, heart rate variability, or conductivity were detected in SARS-CoV-2 inoculated mice of either sex. When re-inoculated 6 months after the first inoculation, hACE2/hTMPRSS2 KI became re-infected with disease signs similar to after the first inoculation. Together these data show that a newly generated hACE2/hTMPRSS2 KI mouse can be used to study mild COVID-19.
Collapse
Affiliation(s)
- Hongwei Liu
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, United States
| | - Terza Brostoff
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, United States
| | - Ana Ramirez
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, United States
| | - Talia Wong
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, United States
| | - Douglas J. Rowland
- Center for Molecular and Genomic Imaging, College of Engineering, University of California, Davis, Davis, CA, United States
| | - Mollie Heffner
- Mouse Biology Program, University of California, Davis, Davis, CA, United States
| | - Arturo Flores
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, United States
| | - Brandon Willis
- Mouse Biology Program, University of California, Davis, Davis, CA, United States
| | - Jeffrey J. Evans
- Mouse Biology Program, University of California, Davis, Davis, CA, United States
| | - Louise Lanoue
- Mouse Biology Program, University of California, Davis, Davis, CA, United States
| | - K. C. Kent Lloyd
- Mouse Biology Program, University of California, Davis, Davis, CA, United States
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Lark L. Coffey
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, United States
| |
Collapse
|
3
|
Hsia J, Spyropoulos AC, Piazza G, Weng S, Dunne MW, Lipardi C, Barnathan ES, Bonaca MP. Antithrombotic Prophylaxis with Rivaroxaban in Patients with Prehospital COVID-19: A Meta-analysis of Two Placebo-Controlled Trials. Thromb Haemost 2024; 124:649-655. [PMID: 37995748 PMCID: PMC11199042 DOI: 10.1055/a-2216-5848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/21/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND We conducted a prespecified meta-analysis of two randomized, placebo-controlled trials of rivaroxaban 10 mg daily in prehospital patients with acute coronavirus disease 2019 (COVID-19). Individually, the trials had limited power to detect a treatment effect due to recruitment stopping ahead of plan. MATERIAL AND METHODS The statistical analysis plan for the meta-analysis was finalized before unblinding of PREVENT-HD, the larger of the two trials. Pooled risk ratios and pooled risk differences along with the two-sided 95% confidence intervals were calculated using random-effect models. RESULTS Rivaroxaban did not reduce the occurrence of either the primary prespecified endpoint, a composite of symptomatic arterial and venous thromboembolism, myocardial infarction, ischemic stroke, acute limb ischemia, all-cause hospitalization, and all-cause mortality (risk difference: 0.0044; 95% confidence interval: -0.0263, 0.0175; p = 0.69 for pooled risk difference) or the secondary endpoint of all-cause hospitalization (p = 0.76). Although thrombotic events were infrequent, pooled analysis did reveal that rivaroxaban reduced arterial and venous thrombotic events (placebo 6 events, rivaroxaban 0 events; pooled risk difference: -0.0068; 95% confidence interval: -0.0132, -0.0006; p = 0.03). In the pooled studies, only one major bleeding event was observed in a rivaroxaban-allocated patient with no critical site or fatal bleeding events. CONCLUSION Although this meta-analysis does not support antithrombotic prophylaxis with rivaroxaban in a broad prehospital population with acute COVID-19, the prevention of arterial and venous thrombotic events among rivaroxaban-allocated patients is consistent with the known thromboprophylactic effect of the drug in medically ill patients.
Collapse
Affiliation(s)
- Judith Hsia
- Colorado Prevention Center and Department of Medicine, University of Colorado, Aurora, Colorado, United States
| | - Alex C. Spyropoulos
- Feinstein Institutes for Medical Research and Department of Medicine, Northwell Health, New York, New York, United States
| | - Gregory Piazza
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States
| | - Stephen Weng
- Janssen Research and Development, Statistical Decision Sciences, Cardiovascular and Metabolism, High Wycombe, United Kingdom
| | - Michael W. Dunne
- Bill and Melinda Gates Medical Research Institute, Cambridge, Massachusetts, United States
| | - Concetta Lipardi
- Janssen Research and Development, Clinical Development, Raritan, New Jersey, United States
| | - Elliot S. Barnathan
- Janssen Research and Development, Clinical Development, Raritan, New Jersey, United States
| | - Marc P. Bonaca
- Colorado Prevention Center and Department of Medicine, University of Colorado, Aurora, Colorado, United States
| |
Collapse
|
4
|
Glazier CR, Baciewicz FA. Epidemiology, Etiology, and Pathophysiology of Pulmonary Embolism. Int J Angiol 2024; 33:76-81. [PMID: 38846994 PMCID: PMC11152621 DOI: 10.1055/s-0044-1785487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024] Open
Abstract
Over the last 20 years, there has been a progressive increase in the incidence of pulmonary embolism (PE) diagnosis in the United States, Europe, and Australia. Increased use of computed tomography pulmonary angiography has likely contributed in part to this rising incidence. However, it is pertinent to note that the burden of comorbidities associated with PE, such as malignancy, obesity, and advanced age, has also increased over the past 20 years. Time-trend analysis in North American, European, and Asian populations suggests that mortality rates associated with PE have been declining. The reported improved survival rates in PE over the past 20 years are likely, at least in part, to be the result of better adherence to guidelines, improved risk stratification, and enhanced treatment. Factors contributing to the development of venous thromboembolism (VTE) include stasis of blood, hypercoagulability, endothelial injury, and inflammation. In 70 to 80% of cases of PE, the thrombi embolizes from the proximal deep veins of the lower extremities and pelvis. Strong risk factors for VTE include lower extremity fractures and surgeries, major trauma, and hospitalization within the previous 3 months for acute myocardial infarction or heart failure with atrial fibrillation. Acute PE causes several pathophysiological responses including hypoxemia and right ventricle (RV) failure. The latter is a result of pulmonary artery occlusion and associated vasoconstriction. Hemodynamic compromise from RV failure is the principal cause of poor outcome in patients with acute PE.
Collapse
Affiliation(s)
| | - Frank A. Baciewicz
- Division of Cardiothoracic Surgery, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
5
|
Bikdeli B, Krishnathasan D, Khairani CD, Bejjani A, Davies J, Porio N, Tristani A, Armero A, Assi AA, Nauffal V, Campia U, Almarzooq Z, Wei E, Ortiz-Rios MD, Zuluaga-Sánchez V, Achanta A, Jesudasen SJ, Tiu B, Merli GJ, Leiva O, Fanikos J, Grandone E, Sharma A, Rizzo S, Pfeferman MB, Morrison RB, Vishnevsky A, Hsia J, Nehler MR, Welker J, Bonaca MP, Carroll B, Goldhaber SZ, Lan Z, Piazza G. Low absolute risk of thrombotic and cardiovascular events in outpatient pregnant women with COVID-19. Thromb Res 2024; 237:209-215. [PMID: 38677791 DOI: 10.1016/j.thromres.2024.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/17/2024] [Accepted: 02/20/2024] [Indexed: 04/29/2024]
Abstract
INTRODUCTION Pregnancy may contribute to an excess risk of thrombotic or cardiovascular events. COVID-19 increases the risk of these events, although the risk is relatively limited among outpatients. We sought to determine whether outpatient pregnant women with COVID-19 are at a high risk for cardiovascular or thrombotic events. MATERIALS & METHODS We analyzed pregnant outpatients with COVID-19 from the multicenter CORONA-VTE-Network registry. The main study outcomes were a composite of adjudicated venous or arterial thrombotic events, and a composite of adjudicated cardiovascular events. Events were assessed 90 days after the COVID-19 diagnosis and reported for non-pregnant women ≤45 years, and for men ≤45 years, as points of reference. RESULTS Among 6585 outpatients, 169 were pregnant at diagnosis. By 90-day follow-up, two pregnant women during the third trimester had lower extremity venous thrombosis, one deep and one superficial vein thrombosis. The cumulative incidence of thrombotic events was 1.20 % (95 % confidence interval [CI]: 0.0 to 2.84 %). Respective rates were 0.47 % (95 % CI: 0.14 % to 0.79 %) among non-pregnant women, and 0.49 % (95 % CI: 0.06 % to 0.91 %) among men ≤45 years. No non-thrombotic cardiovascular events occurred in pregnant women. The rates of cardiovascular events were 0.53 % (95 % CI: 0.18 to 0.87) among non-pregnant women, and 0.68 % (95 % CI: 0.18 to 1.18) in men aged ≤45 years. CONCLUSIONS Thrombotic and cardiovascular events are rare among outpatients with COVID-19. Although a higher event rate among outpatient pregnant women cannot be excluded, the absolute event rates are low and do not warrant population-wide cardiovascular interventions to optimize outcomes.
Collapse
Affiliation(s)
- Behnood Bikdeli
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA; YNHH/ Yale Center for Outcomes Research and Evaluation (CORE), New Haven, CT, USA; Cardiovascular Research Foundation (CRF), New York, NY, USA
| | - Darsiya Krishnathasan
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Candrika D Khairani
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Antoine Bejjani
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Julia Davies
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Nicole Porio
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Anthony Tristani
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Andre Armero
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Ali A Assi
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Victor Nauffal
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Umberto Campia
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Zaid Almarzooq
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Eric Wei
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Marcos D Ortiz-Rios
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Aditya Achanta
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Sirus J Jesudasen
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Bruce Tiu
- Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Geno J Merli
- Department of Cardiology, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Orly Leiva
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - John Fanikos
- Department of Pharmacy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Elvira Grandone
- Thrombosis and Haemostasis Unit, Fondazione I.R.C.C.S. "Casa Sollievo della Sofferenza", S. Giovanni Rotondo (Foggia), Italy; Department of Obstetrics and Gynecology, First I.M. Sechenov Moscow State Medical University, Moscow, Russia
| | - Aditya Sharma
- Department of Medicine, Cardiovascular Medicine, University of Virginia Health, Charlottesville, VA, USA
| | - Samantha Rizzo
- Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Mariana B Pfeferman
- Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Ruth B Morrison
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Alec Vishnevsky
- Department of Cardiology, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Judith Hsia
- CPC Clinical Research, Aurora, CO, USA; Department of Medicine, University of Colorado, Aurora, CO, USA
| | | | - James Welker
- Anne Arundel Research Institute, Annapolis, MD, USA
| | - Marc P Bonaca
- CPC Clinical Research, Aurora, CO, USA; Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Brett Carroll
- Smith Center for Cardiovascular Outcomes Research, Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Samuel Z Goldhaber
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Zhou Lan
- Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA; Center for Clinical Investigation, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gregory Piazza
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, USA.
| |
Collapse
|
6
|
Violi F, Harenberg J, Pignatelli P, Cammisotto V. COVID-19 and Long-COVID Thrombosis: From Clinical and Basic Science to Therapeutics. Thromb Haemost 2024; 124:286-296. [PMID: 37967846 DOI: 10.1055/s-0043-1776713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
Coronavirus infectious disease-19 (COVID-19) is a pandemic characterized by serious lung disease and thrombotic events in the venous and circulation trees, which represent a harmful clinical sign of poor outcome. Thrombotic events are more frequent in patients with severe disease requiring intensive care units and are associated with platelet and clotting activation. However, after resolution of acute infection, patients may still have clinical sequelae, the so-called long-COVID-19, including thrombotic events again in the venous and arterial circulation. The mechanisms accounting for thrombosis in acute and long COVID-19 have not been fully clarified; interactions of COVID-19 with angiotensin converting enzyme 2 or toll-like receptor family or infection-induced cytokine storm have been suggested to be implicated in endothelial cells, leucocytes, and platelets to elicit clotting activation in acute as well in chronic phase of the disease. In acute COVID-19, prophylactic or full doses of anticoagulants exert beneficial effects even if the dosage choice is still under investigation; however, a residual risk still remains suggesting a need for a more appropriate therapeutic approach. In long COVID-19 preliminary data provided useful information in terms of antiplatelet treatment but definition of candidates for thrombotic prophylaxis is still undefined.
Collapse
Affiliation(s)
- Francesco Violi
- Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
- Mediterranea Cardiocentro, Via Orazio, Naples, Italy
| | - Job Harenberg
- Medical Faculty Mannheim, Ruprecht-karls University Heidelberg, Heidelberg, Germany
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
- Mediterranea Cardiocentro, Via Orazio, Naples, Italy
| | - Vittoria Cammisotto
- Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
7
|
Yamashita Y, Yachi S, Takeyama M, Nishimoto Y, Tsujino I, Nakamura J, Yamamoto N, Nakata H, Ikeda S, Umetsu M, Aikawa S, Hayashi H, Satokawa H, Okuno Y, Iwata E, Ogihara Y, Ikeda N, Kondo A, Iwai T, Yamada N, Ogawa T, Kobayashi T, Mo M. Prophylactic Anticoagulation and Thrombosis in Hospitalized Patients with Clinically Stable COVID-19 at Admission: From the Practice-Based Observational Study. Ann Vasc Dis 2024; 17:1-8. [PMID: 38628927 PMCID: PMC11018098 DOI: 10.3400/avd.oa.23-00031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 10/29/2023] [Indexed: 04/19/2024] Open
Abstract
Objectives: The potential benefit of routine prophylactic anticoagulation for all hospitalized patients with clinically stable coronavirus disease 2019 (COVID-19) is still controversial. Method: The CLOT-COVID Study was a multicenter observational study enrolling 2894 consecutive hospitalized patients with COVID-19. The current study population consisted of 1738 hospitalized patients with mild COVID-19 at admission not requiring oxygen administration, who were divided into 2 groups: patients with prophylactic anticoagulation (n = 326) and those without (n = 1412). Results: Patients with prophylactic anticoagulation had more severe status of the worst severity of COVID-19 during hospitalization compared with those without (mild: 38% versus 82%, moderate: 55% versus 17%, and severe or death at discharge: 6.4% versus 0.7%, P <0.001). During hospitalization, 8 patients (0.5%) developed thrombosis, and the incidences of thrombosis were numerically higher in patients with more severe status of worst severity of COVID-19 during hospitalization (mild: 0.2%, moderate: 1.2%, and severe or death at discharge: 3.2%). Conclusions: Among hospitalized patients with clinically stable COVID-19 at admission, patients who did not worsen in COVID-19 severity after admission rarely developed thrombosis, although patients with worsening of COVID-19 severity after admission more often received prophylactic anticoagulation and might have a higher risk of thrombosis.
Collapse
Affiliation(s)
- Yugo Yamashita
- Department of Cardiovascular Medicine, Kyoto University Hospital, Kyoto, Kyoto, Japan
| | - Sen Yachi
- Japan Community Health Care Organization Tokyo Shinjuku Medical Center, Tokyo, Japan
| | - Makoto Takeyama
- Japan Community Health Care Organization Tokyo Shinjuku Medical Center, Tokyo, Japan
| | - Yuji Nishimoto
- Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Hyogo, Japan
| | | | | | | | - Hiroko Nakata
- Yokosuka General Hospital Uwamachi, Yokosuka, Kanagawa, Japan
| | - Satoshi Ikeda
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | | | - Shizu Aikawa
- Tsukuba Medical Center Hospital, Tsukuba, Ibaraki, Japan
| | - Hiroya Hayashi
- Osaka Metropolitan University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hirono Satokawa
- Fukushima Medical University, School of Medicine, Fukushima, Fukushima, Japan
| | - Yoshinori Okuno
- Department of Cardiovascular Medicine, Kyoto University Hospital, Kyoto, Kyoto, Japan
| | - Eriko Iwata
- Nankai Medical Center Japan Community Health Care Organization, Saiki, Oita, Japan
| | | | | | - Akane Kondo
- Shikoku Medical Center for Children and Adults, Zentsuji, Kagawa, Japan
| | | | | | | | | | - Makoto Mo
- Yokohama Minami Kyosai Hospital, Yokohama, Kanagawa, Japan
| | - on behalf of the Clot-COVID Study Investigators
- Department of Cardiovascular Medicine, Kyoto University Hospital, Kyoto, Kyoto, Japan
- Japan Community Health Care Organization Tokyo Shinjuku Medical Center, Tokyo, Japan
- Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Hyogo, Japan
- Hokkaido University Hospital, Sapporo, Hokkaido, Japan
- Hamamatsu Medical Center, Hamamatsu, Shizuoka, Japan
- Yokosuka General Hospital Uwamachi, Yokosuka, Kanagawa, Japan
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
- Tohoku University Hospital, Sendai, Miyagi, Japan
- Tsukuba Medical Center Hospital, Tsukuba, Ibaraki, Japan
- Osaka Metropolitan University Graduate School of Medicine, Suita, Osaka, Japan
- Fukushima Medical University, School of Medicine, Fukushima, Fukushima, Japan
- Nankai Medical Center Japan Community Health Care Organization, Saiki, Oita, Japan
- Mie University Hospital, Tsu, Mie, Japan
- Toho University Ohashi Medical Center, Tokyo, Japan
- Shikoku Medical Center for Children and Adults, Zentsuji, Kagawa, Japan
- Tsukuba Vascular Center, Moriya, Ibaraki, Japan
- Kuwana City Medical Center, Kuwana, Mie, Japan
- Fukushima Daiich Hospital, Fukushima, Fukushima, Japan
- Yokohama Minami Kyosai Hospital, Yokohama, Kanagawa, Japan
| |
Collapse
|
8
|
Zheng R, Solomon A, DiLorenzo M, Rajendran I, Park J, Dhongade V, Garcia MA, Eberhardt RT, Sloan JM, Weinberg J, Klings ES. The Association of Anticoagulation Intensity with Outcomes in Hospitalized COVID-19 Patients. Adv Hematol 2024; 2024:8838308. [PMID: 38500844 PMCID: PMC10948223 DOI: 10.1155/2024/8838308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/20/2024] Open
Abstract
Venous thromboembolism (VTE) risk is increased in patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A key question was whether increased intensity of anticoagulation would help prevent VTE and improve patient outcomes, including transfer to the intensive care unit (ICU) and mortality. At the start of the coronavirus disease-19 (COVID-19) pandemic, our institution, Boston Medical Center, instituted a VTE risk stratification protocol based on patients' initial D-dimer levels, medical history, and presence of thrombosis to determine whether they should receive standard-dose prophylaxis, high-dose prophylaxis, or therapeutic anticoagulation. We performed a retrospective observational cohort study examining the association of degree of anticoagulation with outcomes in 915 hospitalized COVID-19 patients hospitalized initially on the general inpatient wards between March 1,, 2020, and June 1, 2020. Patients directly hospitalized in the ICU were excluded. Most, 813 patients (89%), in our cohort were on standard-dose prophylaxis; 32 patients (3.5%) received high-dose prophylaxis; 70 patients (7.7%), were treated with therapeutic anticoagulation. VTE occurred in 45 patients (4.9%), and the overall in-hospital mortality rate was 5.4% (49 deaths). On multivariable analysis of clinical outcomes in relation to type of anticoagulation, in the high-dose prophylaxis group, there was a trend towards increased in-hospital mortality (odds ratio 2.4 (0.8-7.5, 95% CI)) and increased ICU transfer (odds ratio 2.2 (0.9-5.7, 95% CI)). Our results suggest that patients receiving high-dose prophylaxis had more severe disease that was not mitigated by intermediate-dose anticoagulation.
Collapse
Affiliation(s)
- Rena Zheng
- UMass Chan Medical School, UMass Medical Center, Department of Medicine, Division of Hematology-Oncology, Worcester, MA, USA
| | | | - Madeline DiLorenzo
- New York University Grossman School of Medicine, Department of Medicine, Division of Infectious Diseases and Immunology, New York, NY, USA
| | - Iniya Rajendran
- University of Arizona, College of Medicine, Department of Medicine, Division of Cardiology, Tucson, AZ, USA
| | - Joseph Park
- Brigham and Women's Hospital, Department of Medicine, Boston, MA, USA
| | - Vrushali Dhongade
- Brigham and Women's Hospital, Department of Neurology, Boston, MA, USA
| | - Michael A. Garcia
- Valley Medical Center Pulmonary & Sleep Disorder Clinic, Covington, WA, USA
| | - Robert T. Eberhardt
- Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Department of Medicine, Section of Cardiovascular Medicine, Boston, MA, USA
| | - John Mark Sloan
- Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Department of Medicine, Section of Hematology & Medical Oncology, Boston, MA, USA
| | - Janice Weinberg
- Boston University School of Public Health, Department of Biostatistics, Boston, MA, USA
| | - Elizabeth S. Klings
- Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Department of Medicine, The Pulmonary Center, Boston, MA, USA
| |
Collapse
|
9
|
Boyarchuk O, Perestiuk V, Kosovska T, Volianska L. Coagulation profile in hospitalized children with COVID-19: pediatric age dependency and its impact on long COVID development. Front Immunol 2024; 15:1363410. [PMID: 38510249 PMCID: PMC10950941 DOI: 10.3389/fimmu.2024.1363410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 02/23/2024] [Indexed: 03/22/2024] Open
Abstract
Introduction Pulmonary endotheliopathy and microvascular immunothrombosis play a key role in acute COVID-19. Moreover, persistent endotheliopathy and heightened coagulability frequently occur in individuals recovering from COVID-19, suggesting the intriguing possibility of their role in the development of long COVID. The aim of our study was to investigate the coagulation profile in patients with COVID-19 based on age and their role in the development of long COVID. Methods We conducted a prospective single-center cohort study from September 2022 to August 2023. The study involved 190 patients younger than 18 years who were hospitalized at the Ternopil City Children's Hospital, Ukraine due to COVID-19. Patients underwent determination of coagulation profile in addition to the general clinical examination. After discharge from the hospital, patients were monitored for the presence of long COVID symptoms. Among the 157 participants who consented for follow-up, 62 patients (39.5%) had long COVID symptoms according to the WHO definition, while the rest (95 patients) did not have symptoms of long COVID (fully recovered). Results The study revealed the normal count of platelets in the majority of patients (86.8%), whereas abnormalities in the coagulation profile were revealed in 94.5% of children with COVID-19, and these changes were age-dependent. The patients were mostly presented with increased activated partial thromboplastin time (69.1%), prothrombin time (PT) (39.8%) and D-dimer (45.0%). There was no significant difference between the median of platelet levels and coagulation profile indicators between the groups with long COVID and recovered. Among children who developed persistent long COVID symptoms there was a statistically higher percentage of abnormal PT values (53% versus 36.1%, p=0.0432), with no significant differences in other coagulation profile indicators. Abnormal PT along with female gender, comorbidities, especially allergic pathology, nutritional disorder, including obesity, were determined as potential risk factors of the long COVID development (Odds ratio - 2.0611; 95% 1.0179-4.1737, p=0.0445). Conclusions The study highlights the need for more extensive research into the coagulation profiles of pediatric populations, considering age-specific factors. This could enhance our understanding of thromboinflammation in COVID-19 and its potential contribution to the development of persistent symptoms.
Collapse
|
10
|
Park HS, Lee NK, Lee CS, Byeon SH, Kim SS, Lee SW, Kim YJ. Retinal Artery and Vein Occlusion Risks after Coronavirus Disease 2019 or Coronavirus Disease 2019 Vaccination. Ophthalmology 2024; 131:322-332. [PMID: 37739232 DOI: 10.1016/j.ophtha.2023.09.019] [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: 05/04/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/24/2023] Open
Abstract
PURPOSE To evaluate the incidence of new retinal artery occlusion (RAO) and retinal vein occlusion (RVO) after the diagnosis of coronavirus disease 2019 (COVID-19) or vaccination against COVID-19 and compare the incidences with the population with neither. DESIGN Nationwide population-based cohort study. PARTICIPANTS From a nationwide population-based cohort, 8 418 590 patients were categorized into control (group 1), COVID-19 infection (group 2), and COVID-19 vaccination (group 3) groups. METHODS The cumulative incidence of RAO and RVO was calculated in groups 1, 2, and 3 using the Kaplan-Meier method. We calculated hazard ratios (HRs) and 95% confidence intervals (CIs) based on the Poisson distribution for RAO and RVO according to each group and subgroup using Cox proportional hazards models, with group 1 as the reference. We conducted univariable and multivariable analyses for the risk factors of RAO and RVO according to each subgroup. MAIN OUTCOME MEASURES Cumulative incidence and risks of incidence of RAO and RVO from the index date to day 60. RESULTS In multivariable analysis, no significant increase in RAO and RVO risks after COVID-19 or COVID-19 vaccination were observed in either men or women. These results were observed consistently across various conditions in sensitivity analyses. In subgroup analysis, individuals who were vaccinated before infection showed no significant increase in RAO or RVO risks in both sexes compared with the control group. In the subgroup analysis of vaccinated patients, the HRs of RAO and RVO for different vaccine types did not show an increase compared with the control group; however, an exception was observed in women who received mRNA-1273 vaccines, who showed a higher RAO HR (4.65; 95% CI, 1.27-17.03; P = 0.021). CONCLUSIONS Within 60 days of COVID-19 diagnosis or vaccination, RAO and RVO occurred rarely. We observed no increase in the HR of RVO and RAO relative to COVID-19 or COVID-19 vaccination except for a possible increase in the RAO HR in women who received mRNA-1273, for which the raw incidence was extremely low. Further investigation is required to validate this result. FINANCIAL DISCLOSURE(S) The author(s) have no proprietary or commercial interest in any materials discussed in this article.
Collapse
Affiliation(s)
- Hyo Song Park
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Cheonan, South Korea; Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Nang Kyeong Lee
- Department of Computer Science and Engineering, Sungkyunkwan University, Suwon, South Korea
| | - Christopher Seungkyu Lee
- The Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, South Korea
| | - Suk Ho Byeon
- The Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Soo Kim
- The Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung Won Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, South Korea.
| | - Yong Joon Kim
- The Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
11
|
Gu J, Wang Y, Zhang JF, Wang CQ. The impacts of prophylactic anticoagulation therapy during hospitalization on long-term cardiovascular outcomes in high-risk COVID-19 patients amid the omicron wave of the pandemic. IJC HEART & VASCULATURE 2024; 50:101353. [PMID: 38347941 PMCID: PMC10859301 DOI: 10.1016/j.ijcha.2024.101353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 01/30/2024] [Indexed: 02/15/2024]
Abstract
Background Although prophylactic anticoagulation therapy is suggested to be adopted in severe COVID-19 patients, its effects on the long-term cardiovascular (CV) outcomes, namely the risk of major adverse CV events(MACEs) in high-risk CV patients amid the omicron wave of the pandemic, remain unknown. Methods We conducted this prospective cohort study of consecutive adults hospitalized COVID-19 between 19 April and 12 June 2022, COVID-19 patients with at least two CV risk factors or pre-existing CV diseases were enrolled. A propensity score matching(PSM) method was used to evaluated the effects of prophylactic anticoagulation therapy in hospital on long-term MACEs, including CV death, non-fatal myocardial infarction, non-fatal stroke, hospitalization due to unstable angina pectoris, coronary revascularization and arterial or venous thrombosis. Results Two cohorts (with or without anticoagulants during hospitalization) of each 230 patients with balanced baseline characteristics were formed using PSM. During the 15-month follow-up period, 13 patients with anticoagulants and 29 patients without anticoagulants developed MACEs. Overall, the anticoagulation group had a significantly lower risk of MACEs than the control group (hazard ratio [HR] 0.431; 95 % confidence interval [CI]: 0.224-0.830, P = 0.010). Regarding specific constituents of MACEs, the differences were mainly reflected in arterial or venous thrombosis. The significantly lower HRs of overall MACEs were significantly observed in subgroup of age > 75 years, women, higher D dimer level, unvaccinated and non-nirmatrelvir-ritonavir prescribed patients. Conclusions Prophylactic anticoagulation therapy during hospitalization was effective in reducing long-term MACEs among COVID-19 patients with CV risk factors or pre-existing CV diseases amid the omicron wave of the pandemic.
Collapse
Affiliation(s)
- Jun Gu
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Yue Wang
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Jun-feng Zhang
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Chang-qian Wang
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
12
|
Kamidani R, Okada H, Kawasaki Y, Shimada T, Tamaoki Y, Nakashima Y, Nishio A, Fukuda H, Minamiyama T, Yoshida T, Yoshimura G, Miura T, Ishihara T, Fukuta T, Kitagawa Y, Suzuki K, Miyake T, Doi T, Yoshida T, Tetsuka N, Yoshida S, Ogura S. Impact of augmented renal clearance on anticoagulant therapy in critically ill patients with coronavirus disease 2019: A retrospective cohort study. J Infect Chemother 2024; 30:111-117. [PMID: 37717607 DOI: 10.1016/j.jiac.2023.09.017] [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: 06/18/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 09/19/2023]
Abstract
INTRODUCTION This study aimed to determine the impact of augmented renal clearance (ARC) on anticoagulation therapy in critically ill patients with coronavirus disease 2019 (COVID-19). METHODS This retrospective cohort study included adult patients with severe COVID-19 with ARC who had been treated at our hospital between 2020 and 2021. We measured the estimated glomerular filtration rate calculated by the Chronic Kidney Disease Epidemiology Collaboration formula (eGFRCKD-EPI) every morning, and ARC condition was defined as eGFRCKD-EPI ≥ 130 mL/min/1.73 m2. Multivariate regression analysis with Huber-White sandwich estimator was performed to examine the association of unfractionated heparin (UH) dosage between blood test timings with activated partial thromboplastin time (APTT) compared with and without ARC. RESULTS We identified 38 enrolled patients: seven and 31 in the ARC and non-ARC groups, respectively. In the ARC coexisting condition, a higher dose of UH, which corresponded to the total dose in 24 h from the previous day, was required to achieve the same APTT prolongation, with a significant difference (p < 0.001). CONCLUSIONS Our study suggests that careful monitoring and consideration of higher UH doses in critically ill patients with COVID-19 is necessary because anticoagulation failure can occur during ARC.
Collapse
Affiliation(s)
- Ryo Kamidani
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan; Abuse Prevention Center, Gifu University Graduate School of Medicine, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Hideshi Okada
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan; Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Yuki Kawasaki
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Takuto Shimada
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Yuto Tamaoki
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Yusuke Nakashima
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Ayane Nishio
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Hirotsugu Fukuda
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Toru Minamiyama
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Tomoki Yoshida
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Genki Yoshimura
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Tomotaka Miura
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan; Department of Infection Control, Gifu University Graduate School of Medicine, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Takuma Ishihara
- Innovative and Clinical Research Promotion Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Tetsuya Fukuta
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Yuichiro Kitagawa
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Kodai Suzuki
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Takahito Miyake
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Tomoaki Doi
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Takahiro Yoshida
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Nobuyuki Tetsuka
- Department of Infection Control, Gifu University Graduate School of Medicine, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Shozo Yoshida
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan; Abuse Prevention Center, Gifu University Graduate School of Medicine, Yanagido 1-1, Gifu City, Gifu, Japan.
| | - Shinji Ogura
- Advanced Critical Care Center, Gifu University Hospital, Yanagido 1-1, Gifu City, Gifu, Japan.
| |
Collapse
|
13
|
Pradelle A, Mainbourg S, Provencher S, Massy E, Grenet G, Lega JC. Deaths induced by compassionate use of hydroxychloroquine during the first COVID-19 wave: an estimate. Biomed Pharmacother 2024; 171:116055. [PMID: 38171239 DOI: 10.1016/j.biopha.2023.116055] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND During the first wave of COVID-19, hydroxychloroquine (HCQ) was used off-label despite the absence of evidence documenting its clinical benefits. Since then, a meta-analysis of randomised trials showed that HCQ use was associated with an 11% increase in the mortality rate. We aimed to estimate the number of HCQ-related deaths worldwide. METHODS AND FINDINGS We estimated the worldwide in-hospital mortality attributable to HCQ use by combining the mortality rate, HCQ exposure, number of hospitalised patients, and the increased relative risk of death with HCQ. The mortality rate in hospitalised patients for each country was calculated using pooled prevalence estimated by a meta-analysis of published cohorts. The HCQ exposure was estimated using median and extreme estimates from the same systematic review. The number of hospitalised patients during the first wave was extracted from dedicated databases. The systematic review included 44 cohort studies (Belgium: k = 1, France: k = 2, Italy: k = 12, Spain: k = 6, Turkey: k = 3, USA: k = 20). HCQ prescription rates varied greatly from one country to another (range 16-84%). Overall, using median estimates of HCQ use in each country, we estimated that 16,990 HCQ-related in-hospital deaths (range 6267-19256) occurred in the countries with available data. The median number of HCQ-related deaths in Belgium, Turkey, France, Italy, Spain, and the USA was 240 (range not estimable), 95 (range 92-128), 199 (range not estimable), 1822 (range 1170-2063), 1895 (range 1475-2094) and 12739 (3244- 15570), respectively. CONCLUSIONS Although our estimates are limited by their imprecision, these findings illustrate the hazard of drug repurposing with low-level evidence.
Collapse
Affiliation(s)
- Alexiane Pradelle
- Univ Lyon, Université Claude Bernard University of Lyon 1, Equipe Evaluation et Modélisation des Effets Thérapeutiques, Laboratoire de Biométrie et Biologie Évolutive, UMR CNRS 5558 LBBE, Lyon F-69100, France
| | - Sabine Mainbourg
- Univ Lyon, Université Claude Bernard University of Lyon 1, Equipe Evaluation et Modélisation des Effets Thérapeutiques, Laboratoire de Biométrie et Biologie Évolutive, UMR CNRS 5558 LBBE, Lyon F-69100, France; Unité bases de données cliniques et épidémiologiques, Hospices Civils de Lyon, Lyon F-69310, France; Lyon Immunopathology Federation (LIFe), Hospices Civils de Lyon, Lyon F69000, France
| | - Steeve Provencher
- Pulmonary Hypertension Research Group (http://phrg.ca), Institut universitaire de cardiologie et de pneumologie de Québec Research Center, Department of medicine, Université Laval, Québec City, Canada
| | - Emmanuel Massy
- Lyon Immunopathology Federation (LIFe), Hospices Civils de Lyon, Lyon F69000, France; Service de rhumatologie, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite F69000, France
| | - Guillaume Grenet
- Univ Lyon, Université Claude Bernard University of Lyon 1, Equipe Evaluation et Modélisation des Effets Thérapeutiques, Laboratoire de Biométrie et Biologie Évolutive, UMR CNRS 5558 LBBE, Lyon F-69100, France; Service hospitalo-universitaire de pharmacotoxicologie, Hospices Civils de Lyon, Lyon F69000, France
| | - Jean-Christophe Lega
- Univ Lyon, Université Claude Bernard University of Lyon 1, Equipe Evaluation et Modélisation des Effets Thérapeutiques, Laboratoire de Biométrie et Biologie Évolutive, UMR CNRS 5558 LBBE, Lyon F-69100, France; Lyon Immunopathology Federation (LIFe), Hospices Civils de Lyon, Lyon F69000, France; Service de rhumatologie, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite F69000, France; Service hospitalo-universitaire de pharmacotoxicologie, Hospices Civils de Lyon, Lyon F69000, France.
| |
Collapse
|
14
|
Velásquez PA, Hernandez JC, Galeano E, Hincapié-García J, Rugeles MT, Zapata-Builes W. Effectiveness of Drug Repurposing and Natural Products Against SARS-CoV-2: A Comprehensive Review. Clin Pharmacol 2024; 16:1-25. [PMID: 38197085 PMCID: PMC10773251 DOI: 10.2147/cpaa.s429064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 11/14/2023] [Indexed: 01/11/2024] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a betacoronavirus responsible for the COVID-19 pandemic, causing respiratory disorders, and even death in some individuals, if not appropriately treated in time. To face the pandemic, preventive measures have been taken against contagions and the application of vaccines to prevent severe disease and death cases. For the COVID-19 treatment, antiviral, antiparasitic, anticoagulant and other drugs have been reused due to limited specific medicaments for the disease. Drug repurposing is an emerging strategy with therapies that have already tested safe in humans. One promising alternative for systematic experimental screening of a vast pool of compounds is computational drug repurposing (in silico assay). Using these tools, new uses for approved drugs such as chloroquine, hydroxychloroquine, ivermectin, zidovudine, ribavirin, lamivudine, remdesivir, lopinavir and tenofovir/emtricitabine have been conducted, showing effectiveness in vitro and in silico against SARS-CoV-2 and some of these, also in clinical trials. Additionally, therapeutic options have been sought in natural products (terpenoids, alkaloids, saponins and phenolics) with promising in vitro and in silico results for use in COVID-19 disease. Among these, the most studied are resveratrol, quercetin, hesperidin, curcumin, myricetin and betulinic acid, which were proposed as SARS-CoV-2 inhibitors. Among the drugs reused to control the SARS-CoV2, better results have been observed for remdesivir in hospitalized patients and outpatients. Regarding natural products, resveratrol, curcumin, and quercetin have demonstrated in vitro antiviral activity against SARS-CoV-2 and in vivo, a nebulized formulation has demonstrated to alleviate the respiratory symptoms of COVID-19. This review shows the evidence of drug repurposing efficacy and the potential use of natural products as a treatment for COVID-19. For this, a search was carried out in PubMed, SciELO and ScienceDirect databases for articles about drugs approved or under study and natural compounds recognized for their antiviral activity against SARS-CoV-2.
Collapse
Affiliation(s)
- Paula Andrea Velásquez
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Juan C Hernandez
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Elkin Galeano
- Grupo Productos Naturales Marinos, Departamento de Farmacia, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Jaime Hincapié-García
- Grupo de investigación, Promoción y prevención farmacéutica, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia UdeA, Medellín, Colombia
| | - María Teresa Rugeles
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Wildeman Zapata-Builes
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| |
Collapse
|
15
|
Alahmari A, Krishna G, Jose AM, Qoutah R, Hejazi A, Abumossabeh H, Atef F, Almutiri A, Homoud M, Algarni S, AlAhmari M, Alghamdi S, Alotaibi T, Alwadeai K, Alhammad S, Alahmari M. The long-term effects of COVID-19 on pulmonary status and quality of life. PeerJ 2023; 11:e16694. [PMID: 38144193 PMCID: PMC10749089 DOI: 10.7717/peerj.16694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/28/2023] [Indexed: 12/26/2023] Open
Abstract
Background Few studies have looked at how SARS-CoV-2 affects pulmonary function, exercise capacity, and health-related quality of life over time. The purpose of this study was to evaluate these characteristics in post COVID-19 subjects 1 year after recovery. Methods The study included two groups. The case group included post COVID-19 subjects who had recovered after a year, and the control group included healthy participants who had never tested positive for COVID-19. Results The study screened 90 participants, 42 of whom met the eligibility criteria. The findings revealed that the majority of post COVID-19 subjects had relatively normal lung function 1-year post-recovery. A significant reduction in DLCO (B/P%) was observed in the case group vs. control. The exercise capacity test revealed a clinically significant difference in distance walked and a significant difference in the dyspnea post-walk test in the case group compared to the control group. The case group's health-related quality of life domain scores were significantly affected in terms of energy/fatigue, general health, and physical function. Conclusions The post COVID-19 subjects were shown to have well-preserved lung function after 1 year. However, some degree of impairment in diffusion capacity, exercise capacity, and health-related quality of life remained.
Collapse
Affiliation(s)
- Ayedh Alahmari
- Department of Respiratory Therapy, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Gokul Krishna
- Department of Respiratory Therapy, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Ann Mary Jose
- Department of Respiratory Therapy, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Rowaida Qoutah
- Department of Respiratory Therapy, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Aya Hejazi
- Department of Respiratory Therapy, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Hadeel Abumossabeh
- Department of Respiratory Therapy, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Fatima Atef
- Department of Respiratory Therapy, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Alhanouf Almutiri
- Department of Respiratory Therapy, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Mazen Homoud
- Department of Respiratory Therapy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Saleh Algarni
- Department of Respiratory Therapy, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Mohammed AlAhmari
- Dammam Medical Complex, Eastern Health Cluster, Dammam, Saudi Arabia
- Department of Respiratory Care, Prince Sultan Military College of Health Sciences, Dammam, Saudi Arabia
| | - Saeed Alghamdi
- Clinical Technology Department, Respiratory Care Program, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Tareq Alotaibi
- Department of Respiratory Therapy, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Khalid Alwadeai
- Department of Rehabilitation Science, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Saad Alhammad
- Department of Rehabilitation Science, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mushabbab Alahmari
- Department of Respiratory Therapy, University of Bisha, Bisha, Saudi Arabia
| |
Collapse
|
16
|
Tholin B, Ghanima W, Selle ML, Stavem K. Incidence and determinants of venous thromboembolism over 90 days in hospitalized and nonhospitalized patients with COVID-19. J Intern Med 2023; 294:721-729. [PMID: 37518983 DOI: 10.1111/joim.13706] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
INTRODUCTION COVID-19 is associated with an increased risk of venous thromboembolism (VTE), but there is great variation among reported incidence rates. Most previous studies have focused on hospitalized patients with COVID-19, and only a few reports are from population-based registries. METHODS We studied the 90-day incidence of VTE, associated risk factors and all-cause mortality in hospitalized and nonhospitalized patients with COVID-19 in a nationwide cohort. Data on hospitalizations and outpatient visits were extracted from two national registries with mandatory reporting linked by a unique national identification number carried by all Norwegian residents. We performed Cox proportional hazards regression to determine risk factors for VTE after infection with SARS-CoV-2. RESULTS Our study included 30,495 patients with positive SARS-CoV-2 polymerase chain reaction with a mean (SD) age of 41.9 (17.3) years, and 53% were males. Only 2081 (6.8%) were hospitalized. The 90-day incidence of VTE was 0.3% (95% CI: 0.21-0.33) overall and 2.9% (95% CI: 2.3-3.7) in hospitalized patients. Age (hazard ratio [HR] 1.28 per decade, 95% CI: 1.11-1.48, p < 0.05), history of previous VTE (HR 4.69, 95% CI: 2.34-9.40, p < 0.05), and hospitalization for COVID-19 (HR 23.83, 95% CI: 13.48-42.13, p < 0.05) were associated with risk of VTE. CONCLUSIONS The 90-day incidence of VTE in hospitalized and nonhospitalized patients with COVID-19 was in the lower end compared with previous reports, with considerably higher rates in hospitalized than nonhospitalized patients. Risk factors for VTE were consistent with previously reported studies.
Collapse
Affiliation(s)
- Birgitte Tholin
- Clinic of Internal Medicine, Østfold Hospital, Grålum, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Waleed Ghanima
- Clinic of Internal Medicine, Østfold Hospital, Grålum, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Haematology, Oslo University Hospital, Oslo, Norway
| | - Maria Lie Selle
- Health Services Research Unit, Akershus University Hospital, Lørenskog, Norway
| | - Knut Stavem
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Health Services Research Unit, Akershus University Hospital, Lørenskog, Norway
- Department of Pulmonary Medicine, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
17
|
Liu L, Zhou C, Jiang H, Wei H, Zhou Y, Zhou C, Ji X. Epidemiology, pathogenesis, and management of Coronavirus disease 2019-associated stroke. Front Med 2023; 17:1047-1067. [PMID: 38165535 DOI: 10.1007/s11684-023-1041-7] [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: 05/05/2023] [Accepted: 10/15/2023] [Indexed: 01/03/2024]
Abstract
The Coronavirus disease 2019 (COVID-19) epidemic has triggered a huge impact on healthcare, socioeconomics, and other aspects of the world over the past three years. An increasing number of studies have identified a complex relationship between COVID-19 and stroke, although active measures are being implemented to prevent disease transmission. Severe COVID-19 may be associated with an increased risk of stroke and increase the rates of disability and mortality, posing a serious challenge to acute stroke diagnosis, treatment, and care. This review aims to provide an update on the influence of COVID-19 itself or vaccines on stroke, including arterial stroke (ischemic stroke and hemorrhagic stroke) and venous stroke (cerebral venous thrombosis). Additionally, the neurovascular mechanisms involved in SARS-CoV-2 infection and the clinical characteristics of stroke in the COVID-19 setting are presented. Evidence on vaccinations, potential therapeutic approaches, and effective strategies for stroke management has been highlighted.
Collapse
Affiliation(s)
- Lu Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
| | - Chenxia Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
| | - Huimin Jiang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Huimin Wei
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yifan Zhou
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Chen Zhou
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China.
- Department of Neurology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| | - Xunming Ji
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China.
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China.
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China.
| |
Collapse
|
18
|
Theodorou DJ, Theodorou SJ, Rizos D. Major cardiovascular events after COVID-19, event rates post-vaccination, antiviral or anti-inflammatory therapy, and temporal trends: Rationale and methodology of the CORONA-VTE-Network study. Thromb Res 2023; 232:146-147. [PMID: 37734969 DOI: 10.1016/j.thromres.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023]
Affiliation(s)
| | | | - Dimitrios Rizos
- Department of Internal Medicine, Intensive Care Unit, General Hospital of Ioannina, Greece
| |
Collapse
|
19
|
Landi A, Morici N, Vranckx P, Frigoli E, Bonacchini L, Omazzi B, Tresoldi M, Camponovo C, Moccetti T, Valgimigli M. Edoxaban and/or colchicine in outpatients with COVID-19: rationale and design of the CONVINCE trial. J Cardiovasc Med (Hagerstown) 2023; 24:920-930. [PMID: 37942793 DOI: 10.2459/jcm.0000000000001556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
BACKGROUND An excessive inflammatory response and a hypercoagulable state are not infrequent in patients with coronavirus disease-2019 (COVID-19) and are associated with adverse clinical outcomes. However, the optimal treatment strategy for COVID-19 patients managed in the out-of-hospital setting is still uncertain. DESIGN The CONVINCE (NCT04516941) is an investigator-initiated, open-label, blinded-endpoint, 2 × 2 factorial design randomized trial aimed at assessing two independently tested hypotheses (anticoagulation and anti-inflammatory ones) in COVID-19 patients. Adult symptomatic patients (≥18 years of age) within 7 days from reverse transcription-PCR (RT-PCR) diagnosis of SARS-CoV-2 infection managed at home or in nursery settings were considered for eligibility. Eligible patients fulfilling all inclusion and no exclusion criteria were randomized to edoxaban versus no treatment (anticoagulation hypothesis) and colchicine versus no treatment (anti-inflammatory hypothesis) in a 1 : 1:1 : 1 ratio. The study had two co-primary endpoints (one for each randomization), including the composite of major vascular thrombotic events at 25 ± 3 days for the anticoagulation hypothesis and the composite of SARS-CoV-2 detection rates at 14 ± 3 days by RT-PCR or freedom from death or hospitalizations (anti-inflammatory hypothesis). Study endpoints will be adjudicated by a blinded Clinical Events Committee. With a final sample size of 420 patients, this study projects an 80% power for each of the two primary endpoints appraised separately. CONCLUSION The CONVINCE trial aims at determining whether targeting anticoagulation and/or anti-inflammatory pathways may confer benefit in COVID-19 patients managed in the out-of-hospital setting. TRIAL REGISTRATION ClinicalTrials.gov number, NCT04516941.
Collapse
Affiliation(s)
- Antonio Landi
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale (EOC)
- Department of Biomedical Sciences, University of Italian Switzerland, Lugano, Switzerland
| | - Nuccia Morici
- IRCCS S. Maria Nascente - Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Pascal Vranckx
- the Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Enrico Frigoli
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale (EOC)
| | - Luca Bonacchini
- Emergency Department, ASST Great Metropolitan Hospital Niguarda, Milan
| | - Barbara Omazzi
- Emergency Unit, ASST Rhodense, Garbagnate Milanese, Italy
| | - Moreno Tresoldi
- Unit of General Medicine and Advanced Care, IRCCS San Raffaele Hospital, Milan
| | - Claudio Camponovo
- Department of Anesthesiology, Clinica Ars Medica, Genolier Swiss Medical Network, Gravesano
| | | | - Marco Valgimigli
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale (EOC)
- Department of Biomedical Sciences, University of Italian Switzerland, Lugano, Switzerland
- University of Bern, Bern, Switzerland
| |
Collapse
|
20
|
Park HS, Kim S, Lee CS, Byeon SH, Kim SS, Lee SW, Kim YJ. Retinal vascular occlusion risks during the COVID-19 pandemic and after SARS-CoV-2 infection. Sci Rep 2023; 13:16851. [PMID: 37803163 PMCID: PMC10558568 DOI: 10.1038/s41598-023-44199-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) has been reported to affect vascular networks including the eye. However, evidence on the causal relationship between COVID-19 infection and retinal vascular occlusions remains limited. This study aimed to determine the change in retinal vascular occlusion incidence during COVID-19 era and whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection induces retinal vascular occlusion. Retinal vein occlusion (RVO) and retinal artery occlusion (RAO) incidences during 2018-2019 and 2020-July 2021 were compared, those in confirmed and suspected COVID-19 patients diagnosed from 2020 to January 2021 were calculated, and those in COVID-19 patients during 180 days prior and 180 days after diagnosis were assessed. Additionally, the standardized incidence ratio of RVOs in COVID-19 patients was analyzed. Incidence rates per 100,000 people/year of RVO during 2018-2019 and 2020-2021 was 102.0 and 98.8, respectively. RAO incidence rates during 2018-2019 and 2020-2021 were 11.7 and 12.0, respectively. In both confirmed and suspected COVID-19 patients, the incidence of RVO and RAO did not change significantly from 180 days before to after diagnosis in the adjusted model. RVO incidence slightly decreased while RAO incidence increased during the COVID-19 pandemic. SARS-CoV-2 infection did not significantly increase RVO or RAO incidence.
Collapse
Affiliation(s)
- Hyo Song Park
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Cheonan, South Korea
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Sunyeup Kim
- Department of Medical AI, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Christopher Seungkyu Lee
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Suk Ho Byeon
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Soo Kim
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung Won Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, South Korea.
| | - Yong Joon Kim
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea.
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
21
|
Franco-Mesa C, Walters ET, Shah NR, Palackic A, Wolf SE, Silva MB. Implications of COVID-19 Infection on Arteriovenous Fistula Thrombosis. Vasc Endovascular Surg 2023; 57:732-737. [PMID: 37159054 DOI: 10.1177/15385744231174664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Objective: This study aims to identify and analyze implications of COVID-19 positivity on AVF occlusion, subsequent treatment patterns, and ESRD patient outcomes. Our aim is to provide a quantitative context for vascular access surgeons in order to optimize surgical decision making and minimize patient morbidity. Methods: The de-identified national TriNetX database was queried to extracted all adult patients who had a known AVF between January 1, 2020 and December 31, 2021. From this cohort individuals who also were diagnosed with COVID-19 prior to creation of their AVF were identified. Cohorts were propensity score matched according to age at AVF surgery, gender, ethnicity, diabetes mellitus, nicotine dependence, tobacco use, use of anticoagulant medications, and use of platelet aggregation inhibitors, hypertensive diseases, hyperlipidemia, and prothrombotic states. Results: After propensity score matching there were 5170 patients; 2585 patients in each group. The total patient population had 3023 (58.5%) males and 2147 (41.5%) females. The overall rate of thrombosis of AV fistulas was 300 (11.6%) in the cohort with COVID-19 and 256 (9.9%) in the control group (OR 1.199, CI 1.005-1.43, P =.0453). Open revisions of AVF with thrombectomy were significantly higher in the COVID-19 cohort compared to the non-COVID-19 group (1.5% vs .5% P = .0002, OR 3.199, CI 1.668-6.136). Regarding the time from AVF creation to intervention, the median days for open thrombectomy in COVID-19 patients was 72 vs 105 days in controls. For endovascular thrombectomy, the median was 175 vs 168 days for the COVID-19 and control cohorts respectively. Conclusion: As for this study, there were significant differences in rates of thrombosis and open revisions of recent created AVF, however endovascular interventions remained remarkably low. As noted in this study, the persistent prothrombotic state of patients with a history of COVID-19 may persist beyond the acute infectious period of the disease.
Collapse
Affiliation(s)
- Camila Franco-Mesa
- Department of Surgery, University of Texas Medical Branch, Galveston TX, USA
| | - Elliot T Walters
- Department of Surgery, University of Texas Medical Branch, Galveston TX, USA
| | - Nikhil R Shah
- Department of Surgery, University of Texas Medical Branch, Galveston TX, USA
| | - Alen Palackic
- Department of Surgery, University of Texas Medical Branch, Galveston TX, USA
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Steven E Wolf
- Department of Surgery, University of Texas Medical Branch, Galveston TX, USA
| | - Michael B Silva
- Department of Surgery, University of Texas Medical Branch, Galveston TX, USA
| |
Collapse
|
22
|
Nugraha RA, Muliawan HS, Nugroho NT, Ikhsan M, Adiarto S. Catastrophic Concomitant Arterial and Venous Thrombosis in a Mild COVID-19-Positive Patient. Int J Angiol 2023; 32:197-201. [PMID: 37576528 PMCID: PMC10421675 DOI: 10.1055/s-0041-1735200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Since its first discovery in late 2019, coronavirus disease 2019 (COVID-19) has been a global burden associated with significant morbidity and mortality. COVID-19 has been correlated with the development of hypercoagulable state that predisposes the patients to a higher risk of thromboembolism. Current evidence suggests higher incidence of thrombosis, particularly venous thrombosis, among hospitalized COVID-19 patients, mostly with critical illness. On the other hand, there is currently no data regarding the incidence of vivid thrombosis in ambulatory patients with mild COVID-19 and the incidence of concomitant arterial and venous thrombosis in COVID-19 is extremely rare. Herein, we describe catastrophic outcomes of concomitant lower limb arterial and venous thrombosis in a patient with mild COVID-19. This report highlights the occurrence of concomitant arterial and venous thrombosis in ambulatory setting and that this phenomenon resulted in catastrophic clinical consequences.
Collapse
Affiliation(s)
- Raka A. Nugraha
- Department of General Medicine, Universitas Indonesia Hospital, Depok, Indonesia
| | - Hary S. Muliawan
- Department of Cardiology and Vascular Medicine, National Cardiovascular Center Harapan Kita/Universitas Indonesia Hospital, Depok, Indonesia
| | - Nyityasmono T. Nugroho
- Department of Vascular and Endovascular Surgery, Cipto Mangunkusumo Hospital/Universitas Indonesia Hospital, Depok, Indonesia
| | - Muhammad Ikhsan
- Department of Internal Medicine, Cipto Mangunkusumo Hospital/Universitas Indonesia Hospital, Depok, Indonesia
| | - Suko Adiarto
- Department of Cardiology and Vascular Medicine, National Cardiovascular Center Harapan Kita/Universitas Indonesia Hospital, Depok, Indonesia
| |
Collapse
|
23
|
Baruah P, Patra A, Barge S, Khan MR, Mukherjee AK. Therapeutic Potential of Bioactive Compounds from Edible Mushrooms to Attenuate SARS-CoV-2 Infection and Some Complications of Coronavirus Disease (COVID-19). J Fungi (Basel) 2023; 9:897. [PMID: 37755005 PMCID: PMC10532592 DOI: 10.3390/jof9090897] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 09/28/2023] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a highly infectious positive RNA virus, has spread from its epicenter to other countries with increased mortality and morbidity. Its expansion has hampered humankind's social, economic, and health realms to a large extent. Globally, investigations are underway to understand the complex pathophysiology of coronavirus disease (COVID-19) induced by SARS-CoV-2. Though numerous therapeutic strategies have been introduced to combat COVID-19, none are fully proven or comprehensive, as several key issues and challenges remain unresolved. At present, natural products have gained significant momentum in treating metabolic disorders. Mushrooms have often proved to be the precursor of various therapeutic molecules or drug prototypes. The plentiful bioactive macromolecules in edible mushrooms, like polysaccharides, proteins, and other secondary metabolites (such as flavonoids, polyphenols, etc.), have been used to treat multiple diseases, including viral infections, by traditional healers and the medical fraternity. Some edible mushrooms with a high proportion of therapeutic molecules are known as medicinal mushrooms. In this review, an attempt has been made to highlight the exploration of bioactive molecules in mushrooms to combat the various pathophysiological complications of COVID-19. This review presents an in-depth and critical analysis of the current therapies against COVID-19 versus the potential of natural anti-infective, antiviral, anti-inflammatory, and antithrombotic products derived from a wide range of easily sourced mushrooms and their bioactive molecules.
Collapse
Affiliation(s)
- Paran Baruah
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; (P.B.); (A.P.); (S.B.); (M.R.K.)
- Faculty of Science, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Aparup Patra
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; (P.B.); (A.P.); (S.B.); (M.R.K.)
| | - Sagar Barge
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; (P.B.); (A.P.); (S.B.); (M.R.K.)
| | - Mojibur R. Khan
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; (P.B.); (A.P.); (S.B.); (M.R.K.)
| | - Ashis K. Mukherjee
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; (P.B.); (A.P.); (S.B.); (M.R.K.)
| |
Collapse
|
24
|
Honkanen M, Sirkeoja S, Viskari H, Kailankangas V, Sinisalo M, Syrjänen J. Risk of venous thromboembolism in COVID-19 infection. Intern Med J 2023; 53:1478-1480. [PMID: 37449583 DOI: 10.1111/imj.16145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/25/2023] [Indexed: 07/18/2023]
Abstract
The incidence of venous thromboembolism (VTE) for non-hospitalised patients with coronavirus disease-2019 infection has not been very widely studied. 13 019 persons with a positive SARS-CoV-2 nucleic acid amplification test were identified. In total, 447 (0.2%) VTEs were identified in the study population, 293 (66%) of these were pulmonary embolisms. A positive SARS-CoV-2 test did not increase the risk for VTE in the univariate analysis (odds ratio (OR): 1.0, 95% confidence interval (CI): 0.69-1.4) or multivariable analysis (OR: 1.36, 95% CI: 0.93-1.97).
Collapse
Affiliation(s)
- Meeri Honkanen
- Department of Internal Medicine, Tampere University Hospital and Faculty of Medicine and Life Sciences, Tampere University, Tampere, Finland
| | - Simo Sirkeoja
- Department of Internal Medicine, Tampere University Hospital and Faculty of Medicine and Life Sciences, Tampere University, Tampere, Finland
| | - Hanna Viskari
- Department of Internal Medicine, Tampere University Hospital and Faculty of Medicine and Life Sciences, Tampere University, Tampere, Finland
| | - Ville Kailankangas
- Department of Internal Medicine, Tampere University Hospital and Faculty of Medicine and Life Sciences, Tampere University, Tampere, Finland
| | - Marjatta Sinisalo
- Department of Internal Medicine, Tampere University Hospital and Faculty of Medicine and Life Sciences, Tampere University, Tampere, Finland
| | - Jaana Syrjänen
- Department of Internal Medicine, Tampere University Hospital and Faculty of Medicine and Life Sciences, Tampere University, Tampere, Finland
| |
Collapse
|
25
|
Zhang X, Hao A, Lu Y, Huang W. Deep vein thrombosis and validation of the Caprini risk assessment model in Chinese orthopaedic trauma patients: a multi-center retrospective cohort study enrolling 34,893 patients. Eur J Trauma Emerg Surg 2023; 49:1863-1871. [PMID: 37027013 PMCID: PMC10079483 DOI: 10.1007/s00068-023-02265-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/28/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND The risk of venous thromboembolism among orthopaedic trauma patients is high, but prevalence of deep vein thrombosis (DVT) remains unknown. In addition, the Caprini risk assessment model (RAM) score in orthopaedic trauma patients is undetermined in previous research. This study is aimed to determine the incidence of DVT and then validate the Caprini RAM in orthopaedic trauma patients. METHODS This is a retrospective cohort study enrolling orthopaedic trauma inpatients from seven tertiary and secondary hospitals during a 3-year period (from April 1, 2018 through April 30, 2021). Caprini RAM scores were assessed by experienced nurses on admission. The patients with suspected DVT were verified through duplex ultrasonography by qualified radiologists, and then prospectively followed once a year after discharge. RESULTS In total, 34,893 patients were enrolled in our study. The Caprini RAM identified 45.7% of patients at low risk (Caprini score 0-2), 25.9% at medium risk (3-4), and 28.3% at high risk (5-6), highest risk (7-8), and superhigh risk (> 8). Patients with Caprini score > 5 were likely to be older, female, and with longer length of hospital stay. Moreover, 8695 patients had received ultrasonography to detect DVT. The prevalence of DVT was determined to be 19.0% [95% confidence interval (CI) 18.2-19.9%], which significantly increased with Caprini score. The area under curve of the Caprini RAM for DVT was 0.77 (95% CI 0.76-0.78) with a threshold of 4.5. Furthermore, 6108 patients who had received ultrasonography completed the follow-up. DVT patients had a hazard ratio of 1.75 (95% CI 1.11-2.76; P = 0.005) in the mortality, compared to non-DVT ones. Caprini scores were significantly associated with increase in the mortality [odds ratio (OR) 1.14; 95% CI 1.07-1.21; P < 0.001]; DVT remained an independent effect (OR 1.5; 95% CI 1.02-2.26; P = 0.042). CONCLUSIONS The Caprini RAM may be valid in Chinese orthopaedic trauma patients. Prevalence of DVT and higher Caprini score were significantly associated with increased all-cause mortality among orthopaedic trauma patients after discharge. Further study is warranted to explore the causes of higher mortality in patients with DVT.
Collapse
Affiliation(s)
- Xian Zhang
- Department of Intensive Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Anqi Hao
- Department of Information, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yihan Lu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China.
- National Health Commission Key Laboratory of Health Technology Assessment (Fudan University), Shanghai, 200032, China.
| | - Weifeng Huang
- Department of Intensive Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
26
|
Bikdeli B, Khairani CD, Krishnathasan D, Bejjani A, Armero A, Tristani A, Davies J, Porio N, Assi AA, Nauffal V, Campia U, Almarzooq Z, Wei E, Achanta A, Jesudasen SJ, Tiu BC, Merli GJ, Leiva O, Fanikos J, Sharma A, Vishnevsky A, Hsia J, Nehler MR, Welker J, Bonaca MP, Carroll BJ, Lan Z, Goldhaber SZ, Piazza G. Major cardiovascular events after COVID-19, event rates post-vaccination, antiviral or anti-inflammatory therapy, and temporal trends: Rationale and methodology of the CORONA-VTE-Network study. Thromb Res 2023; 228:94-104. [PMID: 37302267 PMCID: PMC10226776 DOI: 10.1016/j.thromres.2023.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is associated with excess risk of cardiovascular and thrombotic events in the early post-infection period and during convalescence. Despite the progress in our understanding of cardiovascular complications, uncertainty persists with respect to more recent event rates, temporal trends, association between vaccination status and outcomes, and findings within vulnerable subgroups such as older adults (aged 65 years or older), or those undergoing hemodialysis. Sex-informed findings, including results among pregnant and breastfeeding women, as well as adjusted comparisons between male and female adults are similarly understudied. METHODS Adult patients, aged ≥18 years, with polymerase chain reaction-confirmed COVID-19 who received inpatient or outpatient care at the participating centers of the registry are eligible for inclusion. A total of 10,000 patients have been included in this multicenter study, with Brigham and Women's Hospital (Boston, MA) serving as the coordinating center. Other sites include Beth Israel Deaconess Medical Center, Anne Arundel Medical Center, University of Virginia Medical Center, University of Colorado Health System, and Thomas Jefferson University Health System. Data elements will be ascertained manually for accuracy. The two main outcomes are 1) a composite of venous or arterial thrombotic events, and 2) a composite of major cardiovascular events, defined as venous or arterial thrombosis, myocarditis or heart failure with inpatient treatment, new atrial fibrillation/flutter, or cardiovascular death. Clinical outcomes are adjudicated by independent physicians. Vaccination status and time of inclusion in the study will be ascertained for subgroup-specific analyses. Outcomes are pre-specified to be reported separately for hospitalized patients versus those who were initially receiving outpatient care. Outcomes will be reported at 30-day and 90-day follow-up. Data cleaning at the sites and the data coordinating center and outcomes adjudication process are in-progress. CONCLUSIONS The CORONA-VTE-Network study will share contemporary information related to rates of cardiovascular and thrombotic events in patients with COVID-19 overall, as well as within key subgroups, including by time of inclusion, vaccination status, patients undergoing hemodialysis, the elderly, and sex-informed analyses such as comparison of women and men, or among pregnant and breastfeeding women.
Collapse
Affiliation(s)
- Behnood Bikdeli
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; YNHH/Yale Center for Outcomes Research and Evaluation (CORE), New Haven, CT, USA; Cardiovascular Research Foundation (CRF), New York, NY, USA
| | - Candrika D Khairani
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Darsiya Krishnathasan
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Antoine Bejjani
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andre Armero
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anthony Tristani
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Julia Davies
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicole Porio
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ali A Assi
- Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Victor Nauffal
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Umberto Campia
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zaid Almarzooq
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Eric Wei
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Aditya Achanta
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Sirus J Jesudasen
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Bruce C Tiu
- Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Geno J Merli
- Department of Cardiology, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Orly Leiva
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - John Fanikos
- Department of Pharmacy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Aditya Sharma
- Department of Medicine, Cardiovascular Medicine, University of Virginia Health, Charlottesville, VA, USA
| | - Alec Vishnevsky
- Department of Cardiology, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Judith Hsia
- CPC Clinical Research, Aurora, CO, USA; Department of Medicine, University of Colorado, Aurora, CO, USA
| | | | - James Welker
- Anne Arundel Research Institute, Annapolis, MD, USA
| | - Marc P Bonaca
- CPC Clinical Research, Aurora, CO, USA; Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Brett J Carroll
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zhou Lan
- Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Center for Clinical Investigation, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Samuel Z Goldhaber
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gregory Piazza
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Ena J, Valls V. Therapeutic-dose anticoagulation or thromboprophylaxis with low-molecular-weight heparin for moderate Covid-19: meta-analysis of randomized controlled trials. Clin Exp Med 2023; 23:1189-1196. [PMID: 36048371 PMCID: PMC9435420 DOI: 10.1007/s10238-022-00876-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/10/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND We carried out a meta-analysis since there is not enough evidence to recommend for or against therapeutic-dose anticoagulation compared with thromboprophylaxis in noncritically ill patients hospitalized with Covid-19. METHODS We performed a systematic literature search using PubMed, Embase, Cochrane Library, and MedRxiv for randomized trials that included therapeutic-dose with low-molecular-weight heparin (LMW) or thromboprophylaxis with LMW heparin in noncritically ill patients admitted to the hospital with Covid-19. We identified five open-label studies for analysis with a total of 3220 patients. Two independent researchers selected, assessed, and extracted the data in duplicate. The outcomes evaluated were all-cause mortality, progression to invasive mechanical ventilation, incidence of venous thromboembolism, and major bleeding. The studies did not show risk for selection, detection, attrition, or reporting bias. RESULTS Therapeutic-dose anticoagulation with LMW heparin compared with thromboprophylaxis with LMW heparin had no significant effect of all-cause death (risk ratio [RR] 0.85; 95% confidence interval [CI] 0.67-1.07; P = 0.16; I2 = 48%), or progression to invasive mechanical ventilation (RR 0.89; CI 0.73-1.08; P = 0.24; I2: 0%). Therapeutic-dose anticoagulation significantly reduced the risk of venous thromboembolic disease (RR 0.42; 95% CI 0.28-0.62; P = 0.0001; I2 = 0%) [Number needed to treat = 37]. Major bleeding occurred in 1.79% of the patients receiving therapeutic-dose anticoagulation and in 0.97% of those receiving thromboprophylaxis [Number needed to harm 125]. CONCLUSION Therapeutic-dose anticoagulation in noncritically ill patients with Covid-19 could be indicated for patients at high risk of venous thromboembolic disease and low risk of bleeding.
Collapse
Affiliation(s)
- Javier Ena
- Servicio de Medicina Interna, Department of Internal Medicine, Hospital Marina Baixa, Av Jaime Botella Mayor, 7, 03570, Villajoyosa, Alicante, Spain.
| | - Victoria Valls
- Department of Public Health, University Miguel Hernandez, Alicante, Spain
| |
Collapse
|
28
|
Arockiam S, Staniforth B, Kepreotis S, Maznyczka A, Bulluck H. A Contemporary Review of Antiplatelet Therapies in Current Clinical Practice. Int J Mol Sci 2023; 24:11132. [PMID: 37446310 DOI: 10.3390/ijms241311132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Antiplatelet therapy plays a crucial role in a number of cardiovascular disorders. We currently have a range of antiplatelet agents in our armamentarium. In this review, we aim to summarise the common antiplatelet agents currently available, and their use in clinic practice. We not only highlight recent trials exploring antiplatelet therapy in atherosclerotic cardiovascular disease, but also in trials related to transcatheter aortic valve implantation and coronavirus disease 2019. Inevitably, the antithrombotic benefits of these drugs are accompanied by an increase in bleeding complications. Therefore, an individualised approach to weighing each patient's thrombotic risk versus bleeding risk is imperative, in order to improve clinical outcomes.
Collapse
Affiliation(s)
- Sacchin Arockiam
- Yorkshire Heart Centre, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds LS2 3AX, UK
| | - Brittany Staniforth
- Yorkshire Heart Centre, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds LS2 3AX, UK
| | - Sacha Kepreotis
- Yorkshire Heart Centre, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds LS2 3AX, UK
| | - Annette Maznyczka
- Yorkshire Heart Centre, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds LS2 3AX, UK
| | - Heerajnarain Bulluck
- Yorkshire Heart Centre, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds LS2 3AX, UK
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
29
|
Bhoelan S, Codreanu C, Tichelaar V, Borjas Howard J, Meijer K. Exploring heterogeneity in reported venous thromboembolism risk in COVID-19 and comparison to other viral pneumonias: a systematic review and meta-regression. Res Pract Thromb Haemost 2023; 7:102146. [PMID: 37663366 PMCID: PMC10470259 DOI: 10.1016/j.rpth.2023.102146] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/18/2023] [Accepted: 06/24/2023] [Indexed: 09/05/2023] Open
Abstract
Background Sources of heterogeneity in venous thromboembolism (VTE) risk in COVID-19 are unclear and comparisons to other viruses are lacking. Objectives To describe VTE risk in patients with COVID-19, explore sources of heterogeneity, and make comparisons with other viral pneumonia. Methods PubMed and Embase data were searched on March 14, 2021, for studies on VTE in adults hospitalized with viral pneumonia. VTE risk estimates were pooled in a random effects meta-analysis stratified by virus type. Heterogeneity in COVID-19 was explored in multivariable meta-regression. Results Seventy studies in COVID-19 (intensive care [ICU] [47] vs ward [23]), 4 studies in seasonal influenza (ICU [3] vs ward [1]), 2 ICU studies in H1N1 and 1 ICU study in SARS-CoV-1 were included. For COVID-19 ICU, pooled VTE risk was 19.6% (95% confidence interval [CI], 16.2%-23.5; I2 = 92.8%) for nonscreening studies and 30.0% (95% CI, 17.9%-45.7%; I2 = 81.9%) for screening studies. For COVID-19 ward, pooled VTE risk was 3.4% (95% CI, 2.4%-4.7%; I2 = 91.3%) and 22.5% (95% CI, 10.2%-42.7%; I2 = 91.6%) for nonscreening and screening studies, respectively. Higher sample size was associated with lower VTE risk. Pooled VTE risk in seasonal influenza and H1N1 at ICU were 9.0% (95% CI, 5.6%-14.2%; I2 = 39.7%) and 29.2% (95% CI, 8.7%-64.2%; I2 = 77.9%), respectively. At ward, VTE risk of seasonal influenza was 2.4% (95% CI, 2.1%-2.7%). In SARS-CoV-1, VTE risk was 47.8% (95% CI, 34.0-62.0). Conclusion Pooled risk estimates in COVID-19 should be interpreted cautiously as a high degree of heterogeneity is present, which hinders comparison to other viral pneumonia. The association of VTE risk in COVID-19 to sample size suggests publication bias.
Collapse
Affiliation(s)
- Soerajja Bhoelan
- Department of Haematology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Catalina Codreanu
- Department of Haematology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Vladimir Tichelaar
- Department of Haematology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Jaime Borjas Howard
- Department of Haematology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Karina Meijer
- Department of Haematology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
30
|
Bagheri B, Alipour A, Yousefi M, Jalalian R, Moghimi M, Mohammadi M, Hassanpour N, Iranian M. Prevalence of Thromboembolic Events, Including Venous Thromboembolism and Arterial Thrombosis, in Patients with COVID-19: A Systematic Review with Meta-Analysis. J Tehran Heart Cent 2023; 18:154-169. [PMID: 38146412 PMCID: PMC10748660 DOI: 10.18502/jthc.v18i3.14110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 06/23/2023] [Indexed: 12/27/2023] Open
Abstract
Background Many studies have evaluated thromboembolic events in COVID-19 patients, and most of them have reported a high estimation of the prevalence of such events. The present study sought to evaluate the prevalence of thromboembolic events in patients with COVID-19. Methods This study is a systematic review with meta-analysis that investigated thromboembolic events in patients with COVID-19 from the start of the pandemic to August 31, 2021. The 4 main databases for collecting articles were Medline, Scopus, Google Scholar, and Web of Science. Deep vein thrombosis, pulmonary embolism, arterial thrombosis, and the overall rate of thromboembolic events were considered primary outcomes. Results In a total of 63 studies (104 920 patients with COVID-19), the overall thrombosis rate was 21% (95% CI, 18% to 25%), the rate of deep vein thrombosis was 20% (95% Cl, 16% to 25%), the rate of pulmonary embolism was 8% (95% Cl, 6% to 10%), and the rate of arterial thrombosis was 5% (95% Cl, 3% to 7%). The prevalence of all primary outcomes in critically ill patients admitted to the intensive care unit (ICU) was significantly higher (P<0.05). In older patients, the prevalence of overall thrombosis, pulmonary embolism, or deep vein thrombosis was significantly higher (P<0.05). Conclusion This study showed that COVID-19 increases the risk of thromboembolic events, especially in elderly and critically ill patients admitted to the ICU. Therefore, more strategies are needed to prevent thromboembolic events in patients with COVID-19, especially in ICU-admitted and elderly patients.
Collapse
Affiliation(s)
- Babak Bagheri
- Department of Cardiology, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abbas Alipour
- Community Medicine Department, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mojtaba Yousefi
- Department of Cardiology, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Rozita Jalalian
- Department of Cardiology, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Minoo Moghimi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahsa Mohammadi
- Rajaie Cardiovascular Medical and Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Negar Hassanpour
- Rajaie Cardiovascular Medical and Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Iranian
- Rajaie Cardiovascular Medical and Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Piazza G, Spyropoulos AC, Hsia J, Goldin M, Towner WJ, Go AS, Bull TM, Weng S, Lipardi C, Barnathan ES, Bonaca MP. Rivaroxaban for Prevention of Thrombotic Events, Hospitalization, and Death in Outpatients With COVID-19: A Randomized Clinical Trial. Circulation 2023; 147:1891-1901. [PMID: 37154020 PMCID: PMC10270282 DOI: 10.1161/circulationaha.123.063901] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/11/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND COVID-19 (coronavirus disease 2019) is associated with heightened risks of venous and arterial thrombosis and hospitalization due to respiratory failure. To assess whether prophylactic anticoagulation can safely reduce the frequency of venous and arterial thrombosis, hospitalization, and death in nonhospitalized patients with symptomatic COVID-19 and at least one thrombosis risk factor, we conducted the PREVENT-HD double-blind, placebo-controlled randomized trial (A Study of Rivaroxaban to Reduce the Risk of Major Venous and Arterial Thrombotic Events, Hospitalization and Death in Medically Ill Outpatients With Acute, Symptomatic COVID-19] Infection). METHODS PREVENT-HD was conducted between August 2020 and April 2022 at 14 US integrated health care delivery networks. A virtual trial design used remote informed consent and clinical monitoring and facilitated data collection through electronic health record integration with a cloud-based research platform. Nonhospitalized patients with symptomatic COVID-19 and at least one thrombosis risk factor were enrolled and randomly assigned to either 10 mg of oral rivaroxaban or placebo daily for 35 days. The primary efficacy outcome was time to first occurrence of a composite of symptomatic venous thromboembolism, myocardial infarction, ischemic stroke, acute limb ischemia, non-central nervous system systemic arterial embolism, hospitalization, or death through day 35. The principal safety end point was International Society on Thrombosis and Hemostasis critical-site or fatal bleeding. The last study visit was on day 49. RESULTS The study was terminated prematurely because of enrollment challenges and a lower-than-expected blinded pooled event rate. A total of 1284 patients underwent randomization with complete accrual of primary events through May 2022. No patients were lost to follow-up. The primary efficacy outcome occurred in 22 of 641 in the rivaroxaban group and 19 of 643 in the placebo group (3.4% versus 3.0%; hazard ratio, 1.16 [95% CI, 0.63-2.15]; P=0.63). No patient in either group experienced critical-site or fatal bleeding. One patient receiving rivaroxaban had a major bleed. CONCLUSIONS The study was terminated prematurely after enrollment of 32% of planned accrual because of recruitment challenges and lower-than-expected event rate. Rivaroxaban prescribed for 35 days in nonhospitalized patients with symptomatic COVID-19 at risk for thrombosis did not appear to reduce a composite end point of venous and arterial thrombotic events, hospitalization, and death. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT04508023.
Collapse
Affiliation(s)
- Gregory Piazza
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (G.P.)
| | - Alex C. Spyropoulos
- Institute of Health System Science, Feinstein Institutes for Medical Research (A.C.S., M.G.), Northwell Health, Manhasset, NY
- Anticoagulation and Clinical Thrombosis Service (A.C.S.), Northwell Health, Manhasset, NY
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY (A.C.S.)
| | - Judith Hsia
- Colorado Prevention Center Clinical Research, Aurora, CO (J.H., M.P.B.)
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (J.H., M.P.B.)
| | - Mark Goldin
- Institute of Health System Science, Feinstein Institutes for Medical Research (A.C.S., M.G.), Northwell Health, Manhasset, NY
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY (M.G.)
| | - William J. Towner
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena (W.J.T.)
- Department of Infectious Disease, Kaiser Permanente Los Angeles Medical Center, CA (W.J.T.)
- Departments of Clinical Science (W.J.T.), Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA
| | - Alan S. Go
- Health Systems Science (A.S.G.), Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA
- Division of Research, Kaiser Permanente Northern California, Oakland (A.S.G.)
- Departments of Epidemiology, Biostatistics and Medicine, University of California, San Francisco (A.S.G.)
| | - Todd M. Bull
- Pulmonary Sciences and Critical Care Medicine and Cardiology, University of Colorado School of Medicine, Aurora (T.M.B.)
| | - Stephen Weng
- Janssen Research and Development, Statistical Decision Sciences, Cardiovascular and Metabolism, High Wycombe, UK (S.W.)
| | - Concetta Lipardi
- Janssen Research and Development, Clinical Development, Raritan, NJ (C.L., E.S.B.)
| | - Elliot S. Barnathan
- Janssen Research and Development, Clinical Development, Raritan, NJ (C.L., E.S.B.)
| | - Marc P. Bonaca
- Colorado Prevention Center Clinical Research, Aurora, CO (J.H., M.P.B.)
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (J.H., M.P.B.)
| |
Collapse
|
32
|
Piwowarczyk P, Szczukocka M, Cios W, Okuńska P, Raszewski G, Borys M, Wiczling P, Czuczwar M. Population Pharmacokinetics and Probability of Target Attainment Analysis of Nadroparin in Different Stages of COVID-19. Clin Pharmacokinet 2023; 62:835-847. [PMID: 37097604 PMCID: PMC10126531 DOI: 10.1007/s40262-023-01244-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 04/26/2023]
Abstract
BACKGROUND AND OBJECTIVE The risk of thrombotic complications in critical patients with COVID-19 remains extremely high, and multicenter trials failed to prove a survival benefit of escalated doses of low-molecular-weight heparins (nadroparin calcium) in this group. The aim of this study was to develop a pharmacokinetic model of nadroparin according to different stages of COVID-19 severity. METHODS Blood samples were obtained from 43 patients with COVID-19 who received nadroparin and were treated with conventional oxygen therapy, mechanical ventilation, and extracorporeal membrane oxygenation. We recorded clinical, biochemical, and hemodynamic variables during 72 h of treatment. The analyzed data comprised 782 serum nadroparin concentrations and 219 anti-Xa levels. We conducted population nonlinear mixed-effects modeling (NONMEM) and performed Monte Carlo simulations of the probability of target attainment for reaching 0.2-0.5 IU/mL anti-Xa levels in study groups. RESULTS We successfully developed a one-compartment model to describe the population pharmacokinetics of nadroparin in different stages of COVID-19. The absorption rate constant of nadroparin was 3.8 and 3.2 times lower, concentration clearance was 2.22 and 2.93 times higher, and anti-Xa clearance was 0.87 and 1.1 times higher in mechanically ventilated patients and the extracorporeal membrane oxygenation group compared with patients treated with conventional oxygen, respectively. The newly developed model indicated that 5.900 IU of nadroparin given subcutaneously twice daily in the mechanically ventilated patients led to a similar probability of target attainment of 90% as 5.900 IU of subcutaneous nadroparin given once daily in the group supplemented with conventional oxygen. CONCLUSIONS Different nadroparin dosing is required for patients undergoing mechanical ventilation and extracorporeal membrane oxygenation to achieve the same targets as those for non-critically ill patients. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov identifier no. NCT05621915.
Collapse
Affiliation(s)
- Paweł Piwowarczyk
- II Department of Anesthesiology and Intensive Care, Medical University of Lublin, Lublin, Poland
| | - Marta Szczukocka
- II Department of Anesthesiology and Intensive Care, Medical University of Lublin, Lublin, Poland
| | - Wojciech Cios
- Department of Infectious Diseases, Medical University of Lublin, Lublin, Poland
| | - Paulina Okuńska
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Grzegorz Raszewski
- Department of Physiopathology, Institute of Rural Health, Lublin, Poland
| | - Michał Borys
- II Department of Anesthesiology and Intensive Care, Medical University of Lublin, Lublin, Poland
| | - Paweł Wiczling
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Mirosław Czuczwar
- II Department of Anesthesiology and Intensive Care, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
33
|
Baroni C, Potito J, Perticone ME, Orausclio P, Luna CM. How Does Long-COVID Impact Prognosis and the Long-Term Sequelae? Viruses 2023; 15:1173. [PMID: 37243259 PMCID: PMC10222218 DOI: 10.3390/v15051173] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/01/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
CONTEXT We reviewed what has been studied and published during the last 3 years about the consequences, mainly respiratory, cardiac, digestive, and neurological/psychiatric (organic and functional), in patients with COVID-19 of prolonged course. OBJECTIVE To conduct a narrative review synthesizing current clinical evidence of abnormalities of signs, symptoms, and complementary studies in COVID-19 patients who presented a prolonged and complicated course. METHODS A review of the literature focused on the involvement of the main organic functions mentioned, based almost exclusively on the systematic search of publications written in English available on PubMed/MEDLINE. RESULTS Long-term respiratory, cardiac, digestive, and neurological/psychiatric dysfunction are present in a significant number of patients. Lung involvement is the most common; cardiovascular involvement may happen with or without symptoms or clinical abnormalities; gastrointestinal compromise includes the loss of appetite, nausea, gastroesophageal reflux, diarrhea, etc.; and neurological/psychiatric compromise can produce a wide variety of signs and symptoms, either organic or functional. Vaccination is not associated with the emergence of long-COVID, but it may happen in vaccinated people. CONCLUSIONS The severity of illness increases the risk of long-COVID. Pulmonary sequelae, cardiomyopathy, the detection of ribonucleic acid in the gastrointestinal tract, and headaches and cognitive impairment may become refractory in severely ill COVID-19 patients.
Collapse
Affiliation(s)
- Carolina Baroni
- Department of Medicine, Pulmonary Diseases Division, Hospital de Clínicas, University of Buenos Aires, Buenos Aires C1120 AAF, Argentina
| | - Jorge Potito
- Department of Medicine, Pulmonary Diseases Division, Hospital de Clínicas, University of Buenos Aires, Buenos Aires C1120 AAF, Argentina
| | - María Eugenia Perticone
- Department of Medicine, Pulmonary Diseases Division, Hospital de Clínicas, University of Buenos Aires, Buenos Aires C1120 AAF, Argentina
| | - Paola Orausclio
- Department of Radiology, Centro Rossi, Buenos Aires C1035 ABC, Argentina
| | - Carlos Marcelo Luna
- Department of Medicine, Pulmonary Diseases Division, Hospital de Clínicas, University of Buenos Aires, Buenos Aires C1120 AAF, Argentina
| |
Collapse
|
34
|
Stone GW, Farkouh ME, Lala A, Tinuoye E, Dressler O, Moreno PR, Palacios IF, Goodman SG, Esper RB, Abizaid A, Varade D, Betancur JF, Ricalde A, Payro G, Castellano JM, Hung IFN, Nadkarni GN, Giustino G, Godoy LC, Feinman J, Camaj A, Bienstock SW, Furtado RHM, Granada C, Bustamante J, Peyra C, Contreras J, Owen R, Bhatt DL, Pocock SJ, Fuster V. Randomized Trial of Anticoagulation Strategies for Noncritically Ill Patients Hospitalized With COVID-19. J Am Coll Cardiol 2023; 81:1747-1762. [PMID: 36889611 PMCID: PMC9987252 DOI: 10.1016/j.jacc.2023.02.041] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND Prior studies of therapeutic-dose anticoagulation in patients with COVID-19 have reported conflicting results. OBJECTIVES We sought to determine the safety and effectiveness of therapeutic-dose anticoagulation in noncritically ill patients with COVID-19. METHODS Patients hospitalized with COVID-19 not requiring intensive care unit treatment were randomized to prophylactic-dose enoxaparin, therapeutic-dose enoxaparin, or therapeutic-dose apixaban. The primary outcome was the 30-day composite of all-cause mortality, requirement for intensive care unit-level of care, systemic thromboembolism, or ischemic stroke assessed in the combined therapeutic-dose groups compared with the prophylactic-dose group. RESULTS Between August 26, 2020, and September 19, 2022, 3,398 noncritically ill patients hospitalized with COVID-19 were randomized to prophylactic-dose enoxaparin (n = 1,141), therapeutic-dose enoxaparin (n = 1,136), or therapeutic-dose apixaban (n = 1,121) at 76 centers in 10 countries. The 30-day primary outcome occurred in 13.2% of patients in the prophylactic-dose group and 11.3% of patients in the combined therapeutic-dose groups (HR: 0.85; 95% CI: 0.69-1.04; P = 0.11). All-cause mortality occurred in 7.0% of patients treated with prophylactic-dose enoxaparin and 4.9% of patients treated with therapeutic-dose anticoagulation (HR: 0.70; 95% CI: 0.52-0.93; P = 0.01), and intubation was required in 8.4% vs 6.4% of patients, respectively (HR: 0.75; 95% CI: 0.58-0.98; P = 0.03). Results were similar in the 2 therapeutic-dose groups, and major bleeding in all 3 groups was infrequent. CONCLUSIONS Among noncritically ill patients hospitalized with COVID-19, the 30-day primary composite outcome was not significantly reduced with therapeutic-dose anticoagulation compared with prophylactic-dose anticoagulation. However, fewer patients who were treated with therapeutic-dose anticoagulation required intubation and fewer died (FREEDOM COVID [FREEDOM COVID Anticoagulation Strategy]; NCT04512079).
Collapse
Affiliation(s)
- Gregg W Stone
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael E Farkouh
- Peter Munk Cardiac Centre, University of Toronto, Toronto, Ontario, Canada
| | - Anuradha Lala
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Elizabeth Tinuoye
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Pedro R Moreno
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Shaun G Goodman
- St Michael's Hospital, Unity Heath, University of Toronto, Toronto, Ontario, Canada; Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| | | | - Alexandre Abizaid
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | | | | | | | - Gerardo Payro
- Instituto Nacional de Ciencias Medicas y Nutrición Salvador Zubiran, Mexico City, Mexico
| | - José María Castellano
- Centro Integral de Enfermedades Cardiovasculares (CIEC), Hospital Universitario Monterpincipe, Grupo HM Hospitales, Madrid, Spain
| | | | - Girish N Nadkarni
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Division of Data Driven and Digital Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gennaro Giustino
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lucas C Godoy
- Peter Munk Cardiac Centre, University of Toronto, Toronto, Ontario, Canada
| | - Jason Feinman
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Anton Camaj
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Solomon W Bienstock
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Carlos Granada
- CogenTech Medical and Digital Innovation, Mahwah, New Jersey, USA
| | - Jessica Bustamante
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Carlos Peyra
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Johanna Contreras
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ruth Owen
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Deepak L Bhatt
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stuart J Pocock
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Valentin Fuster
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
35
|
van den Berg J, Haslbauer JD, Stalder AK, Romanens A, Mertz KD, Studt JD, Siegemund M, Buser A, Holbro A, Tzankov A. Von Willebrand factor and the thrombophilia of severe COVID-19: in situ evidence from autopsies. Res Pract Thromb Haemost 2023; 7:100182. [PMID: 37333991 PMCID: PMC10192064 DOI: 10.1016/j.rpth.2023.100182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 04/29/2023] [Accepted: 05/08/2023] [Indexed: 06/20/2023] Open
Abstract
Background COVID-19 is accompanied by a hypercoagulable state and characterized by microvascular and macrovascular thrombotic complications. In plasma samples from patients with COVID-19, von Willebrand factor (VWF) levels are highly elevated and predictive of adverse outcomes, especially mortality. Yet, VWF is usually not included in routine coagulation analyses, and histologic evidence of its involvement in thrombus formation is lacking. Objectives To determine whether VWF, an acute-phase protein, is a bystander, ie, a biomarker of endothelial dysfunction, or a causal factor in the pathogenesis of COVID-19. Methods We compared autopsy samples from 28 patients with lethal COVID-19 to those from matched controls and systematically assessed for VWF and platelets by immunohistochemistry. The control group comprised 24 lungs, 23 lymph nodes, and 9 hearts and did not differ significantly from the COVID-19 group in age, sex, body mass index (BMI), blood group, or anticoagulant use. Results In lungs, assessed for platelets by immunohistochemistry for CD42b, microthrombi were more frequent in patients with COVID-19 (10/28 [36%] vs 2/24 [8%]; P = .02). A completely normal pattern of VWF was rare in both groups. Accentuated endothelial staining was found in controls, while VWF-rich thrombi were only found in patients with COVID-19 (11/28 [39%] vs 0/24 [0%], respectively; P < .01), as were NETosis thrombi enriched with VWF (7/28 [25%] vs 0/24 [0%], respectively; P < .01). Forty-six percent of the patients with COVID-19 had VWF-rich thrombi, NETosis thrombi, or both. Trends were also seen in pulmonary draining lymph nodes (7/20 [35%] vs 4/24 [17%]; P = .147), where the overall presence of VWF was very high. Conclusion We provide in situ evidence of VWF-rich thrombi, likely attributable to COVID-19, and suggest that VWF may be a therapeutic target in severe COVID-19.
Collapse
Affiliation(s)
- Jana van den Berg
- Department of Hematology, University Hospital Basel, Basel, Switzerland
| | - Jasmin D Haslbauer
- Department of Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Department of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Anna K Stalder
- Department of Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Anna Romanens
- Department of Oncology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Kirsten D Mertz
- Department of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Jan-Dirk Studt
- Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| | - Martin Siegemund
- Intensive Care Unit, Department of Acute Medicine, University Hospital, Basel, Switzerland
| | - Andreas Buser
- Department of Hematology, University Hospital Basel, Basel, Switzerland
- Regional Blood Transfusion Service, Swiss Red Cross, Basel, Switzerland
| | - Andreas Holbro
- Department of Hematology, University Hospital Basel, Basel, Switzerland
- Regional Blood Transfusion Service, Swiss Red Cross, Basel, Switzerland
| | - Alexandar Tzankov
- Department of Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
36
|
Khan MH, Becker RC. Cardiopulmonary Phenotypes of Post Acute Sequelae of Severe Acute Respiratory Syndrome Coronavirus 2: A Narrative Review. Cardiol Rev 2023; 31:117-127. [PMID: 37036191 DOI: 10.1097/crd.0000000000000429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The acute effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are well known; however, the long-term cardiopulmonary effects are less well characterized. The phenotypic expression of acute infection is heterogeneous, ranging from a complete absence of symptoms to shock, multisystem organ failure, and death. Patients with severe or critical coronavirus disease (COVID-19) who survive their initial illness can require a prolonged period of recovery lasting weeks to months. This specific patient group is part of a larger and even more heterogeneous group of patients who initially experience mild-to-moderate symptoms that fail to resolve over time. Collectively, patients recovering from severe or critical COVID-19 and those who continue to experience symptoms following a lower acuity infection are considered to have Post Acute Sequalae of SARS-CoV-2 infection (PASC). Using prognostic factors like myocardial infarction, myocarditis, pulmonary embolism, acute respiratory distress syndrome, need for mechanical ventilation or extracorporeal membrane oxygenation, and advanced pharmaceutical therapies that primarily occur or are instituted in the acute phase of illness one can begin to develop a taxonomy or corpus of PASC in its varied forms.
Collapse
Affiliation(s)
- Muhammad H Khan
- From the Department of Internal Medicine, University of Cincinnati, Cincinnati, OH
| | - Richard C Becker
- University of Cincinnati Heart, Lung and Vascular Institute, Cincinnati, OH
| |
Collapse
|
37
|
Sherif ZA, Gomez CR, Connors TJ, Henrich TJ, Reeves WB. Pathogenic mechanisms of post-acute sequelae of SARS-CoV-2 infection (PASC). eLife 2023; 12:e86002. [PMID: 36947108 DOI: 10.7554/elife.86002:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/10/2023] [Indexed: 08/28/2024] Open
Abstract
COVID-19, with persistent and new onset of symptoms such as fatigue, post-exertional malaise, and cognitive dysfunction that last for months and impact everyday functioning, is referred to as Long COVID under the general category of post-acute sequelae of SARS-CoV-2 infection (PASC). PASC is highly heterogenous and may be associated with multisystem tissue damage/dysfunction including acute encephalitis, cardiopulmonary syndromes, fibrosis, hepatobiliary damages, gastrointestinal dysregulation, myocardial infarction, neuromuscular syndromes, neuropsychiatric disorders, pulmonary damage, renal failure, stroke, and vascular endothelial dysregulation. A better understanding of the pathophysiologic mechanisms underlying PASC is essential to guide prevention and treatment. This review addresses potential mechanisms and hypotheses that connect SARS-CoV-2 infection to long-term health consequences. Comparisons between PASC and other virus-initiated chronic syndromes such as myalgic encephalomyelitis/chronic fatigue syndrome and postural orthostatic tachycardia syndrome will be addressed. Aligning symptoms with other chronic syndromes and identifying potentially regulated common underlining pathways may be necessary for understanding the true nature of PASC. The discussed contributors to PASC symptoms include sequelae from acute SARS-CoV-2 injury to one or more organs, persistent reservoirs of the replicating virus or its remnants in several tissues, re-activation of latent pathogens such as Epstein-Barr and herpes viruses in COVID-19 immune-dysregulated tissue environment, SARS-CoV-2 interactions with host microbiome/virome communities, clotting/coagulation dysregulation, dysfunctional brainstem/vagus nerve signaling, dysautonomia or autonomic dysfunction, ongoing activity of primed immune cells, and autoimmunity due to molecular mimicry between pathogen and host proteins. The individualized nature of PASC symptoms suggests that different therapeutic approaches may be required to best manage specific patients.
Collapse
Affiliation(s)
- Zaki A Sherif
- Department of Biochemistry & Molecular Biology, Howard University College of Medicine, Washington, District of Columbia, United States
| | - Christian R Gomez
- Division of Lung Diseases, National Institutes of Health (NIH), National Heart, Lung and Blood Institute (NHLBI), Bethesda, United States
| | - Thomas J Connors
- Department of Pediatrics, Division of Critical Care, Columbia University Vagelos College of Physicians and Surgeons and New York - Presbyterian Morgan Stanley Children's Hospital, New York, United States
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California, San Francisco, United States
| | - William Brian Reeves
- Department of Medicine, Joe R. and Teresa Lozano Long School of Medicine, University of Texas, San Antonio, United States
| |
Collapse
|
38
|
Sherif ZA, Gomez CR, Connors TJ, Henrich TJ, Reeves WB. Pathogenic mechanisms of post-acute sequelae of SARS-CoV-2 infection (PASC). eLife 2023; 12:e86002. [PMID: 36947108 PMCID: PMC10032659 DOI: 10.7554/elife.86002] [Citation(s) in RCA: 81] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/10/2023] [Indexed: 03/23/2023] Open
Abstract
COVID-19, with persistent and new onset of symptoms such as fatigue, post-exertional malaise, and cognitive dysfunction that last for months and impact everyday functioning, is referred to as Long COVID under the general category of post-acute sequelae of SARS-CoV-2 infection (PASC). PASC is highly heterogenous and may be associated with multisystem tissue damage/dysfunction including acute encephalitis, cardiopulmonary syndromes, fibrosis, hepatobiliary damages, gastrointestinal dysregulation, myocardial infarction, neuromuscular syndromes, neuropsychiatric disorders, pulmonary damage, renal failure, stroke, and vascular endothelial dysregulation. A better understanding of the pathophysiologic mechanisms underlying PASC is essential to guide prevention and treatment. This review addresses potential mechanisms and hypotheses that connect SARS-CoV-2 infection to long-term health consequences. Comparisons between PASC and other virus-initiated chronic syndromes such as myalgic encephalomyelitis/chronic fatigue syndrome and postural orthostatic tachycardia syndrome will be addressed. Aligning symptoms with other chronic syndromes and identifying potentially regulated common underlining pathways may be necessary for understanding the true nature of PASC. The discussed contributors to PASC symptoms include sequelae from acute SARS-CoV-2 injury to one or more organs, persistent reservoirs of the replicating virus or its remnants in several tissues, re-activation of latent pathogens such as Epstein-Barr and herpes viruses in COVID-19 immune-dysregulated tissue environment, SARS-CoV-2 interactions with host microbiome/virome communities, clotting/coagulation dysregulation, dysfunctional brainstem/vagus nerve signaling, dysautonomia or autonomic dysfunction, ongoing activity of primed immune cells, and autoimmunity due to molecular mimicry between pathogen and host proteins. The individualized nature of PASC symptoms suggests that different therapeutic approaches may be required to best manage specific patients.
Collapse
Affiliation(s)
- Zaki A Sherif
- Department of Biochemistry & Molecular Biology, Howard University College of MedicineWashington, District of ColumbiaUnited States
| | - Christian R Gomez
- Division of Lung Diseases, National Institutes of Health (NIH), National Heart, Lung and Blood Institute (NHLBI)BethesdaUnited States
| | - Thomas J Connors
- Department of Pediatrics, Division of Critical Care, Columbia University Vagelos College of Physicians and Surgeons and New York - Presbyterian Morgan Stanley Children's HospitalNew YorkUnited States
| | - Timothy J Henrich
- Division of Experimental Medicine, University of CaliforniaSan FranciscoUnited States
| | - William Brian Reeves
- Department of Medicine, Joe R. and Teresa Lozano Long School of Medicine, University of TexasSan AntonioUnited States
| |
Collapse
|
39
|
Stotts C, Corrales-Medina VF, Rayner KJ. Pneumonia-Induced Inflammation, Resolution and Cardiovascular Disease: Causes, Consequences and Clinical Opportunities. Circ Res 2023; 132:751-774. [PMID: 36927184 DOI: 10.1161/circresaha.122.321636] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Pneumonia is inflammation in the lungs, which is usually caused by an infection. The symptoms of pneumonia can vary from mild to life-threatening, where severe illness is often observed in vulnerable populations like children, older adults, and those with preexisting health conditions. Vaccines have greatly reduced the burden of some of the most common causes of pneumonia, and the use of antimicrobials has greatly improved the survival to this infection. However, pneumonia survivors do not return to their preinfection health trajectories but instead experience an accelerated health decline with an increased risk of cardiovascular disease. The mechanisms of this association are not well understood, but a persistent dysregulated inflammatory response post-pneumonia appears to play a central role. It is proposed that the inflammatory response during pneumonia is left unregulated and exacerbates atherosclerotic vascular disease, which ultimately leads to adverse cardiac events such as myocardial infarction. For this reason, there is a need to better understand the inflammatory cross talk between the lungs and the heart during and after pneumonia to develop therapeutics that focus on preventing pneumonia-associated cardiovascular events. This review will provide an overview of the known mechanisms of inflammation triggered during pneumonia and their relevance to the increased cardiovascular risk that follows this infection. We will also discuss opportunities for new clinical approaches leveraging strategies to promote inflammatory resolution pathways as a novel therapeutic target to reduce the risk of cardiac events post-pneumonia.
Collapse
Affiliation(s)
- Cameron Stotts
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (C.S., K.J.R).,Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (C.S., V.F.C.-M.).,University of Ottawa Heart Institute, Ottawa, ON, Canada (C.S., K.J.R)
| | - Vicente F Corrales-Medina
- Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (C.S., V.F.C.-M.).,Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (V.F.C-M).,Ottawa Hospital Research Institute, Ottawa, ON, Canada (V.F.C.-M)
| | - Katey J Rayner
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (C.S., K.J.R).,University of Ottawa Heart Institute, Ottawa, ON, Canada (C.S., K.J.R)
| |
Collapse
|
40
|
Asakawa T, Cai Q, Shen J, Zhang Y, Li Y, Chen P, Luo W, Zhang J, Zhou J, Zeng H, Weng R, Hu F, Feng H, Chen J, Huang J, Zhang X, Zhao Y, Fang L, Yang R, Huang J, Wang F, Liu Y, Lu H. Sequelae of long COVID, known and unknown: A review of updated information. Biosci Trends 2023; 17:85-116. [PMID: 36928222 DOI: 10.5582/bst.2023.01039] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Over three years have passed since the COVID-19 pandemic started. The dangerousness and impact of COVID-19 should definitely not be ignored or underestimated. Other than the symptoms of acute infection, the long-term symptoms associated with SARS-CoV-2 infection, which are referred to here as "sequelae of long COVID (LC)", are also a conspicuous global public health concern. Although such sequelae were well-documented, the understanding of and insights regarding LC-related sequelae remain inadequate due to the limitations of previous studies (the follow-up, methodological flaws, heterogeneity among studies, etc.). Notably, robust evidence regarding diagnosis and treatment of certain LC sequelae remain insufficient and has been a stumbling block to better management of these patients. This awkward situation motivated us to conduct this review. Here, we comprehensively reviewed the updated information, particularly focusing on clinical issues. We attempt to provide the latest information regarding LC-related sequelae by systematically reviewing the involvement of main organ systems. We also propose paths for future exploration based on available knowledge and the authors' clinical experience. We believe that these take-home messages will be helpful to gain insights into LC and ultimately benefit clinical practice in treating LC-related sequelae.
Collapse
Affiliation(s)
- Tetsuya Asakawa
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Qingxian Cai
- Department of Hepatology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Jiayin Shen
- Department of Science and Education, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Ying Zhang
- Department of Endocrinology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Yongshuang Li
- Department of Dermatology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Peifen Chen
- Department of Respiratory Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Wen Luo
- Department of Respiratory Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Jiangguo Zhang
- Department of Gastroenterology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Jinfeng Zhou
- Department of Gastroenterology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Hui Zeng
- Department of Cardiology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Ruihui Weng
- Department of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Feng Hu
- Department of Nephrology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Huiquan Feng
- Department of Urology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Jun Chen
- Department of Hepatology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Jie Huang
- Department of Dermatology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Xiaoyin Zhang
- Department of Gastroenterology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Yu Zhao
- Department of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Liekui Fang
- Department of Urology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Rongqing Yang
- Department of Dermatology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Jia Huang
- Department of Intensive Care Unit, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Fuxiang Wang
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Yingxia Liu
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Hongzhou Lu
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China.,Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, China
| |
Collapse
|
41
|
Kommentar zu „Risiko für venöse Thromboembolien bei ambulanten COVID-19-Patienten erhöht“. Dtsch Med Wochenschr 2023. [DOI: 10.1055/a-1962-4659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
|
42
|
Safiriyu I, Fatuyi M, Mehta A, Naseer A, Alexander E, Vovan H, Shamaki GR, Bob-Manuel T. Impact of COVID-19 Infection on the Clinical Outcomes of Pulmonary Embolism Hospitalizations : A Nationwide Analysis. Curr Probl Cardiol 2023; 48:101669. [PMID: 36841316 PMCID: PMC9946871 DOI: 10.1016/j.cpcardiol.2023.101669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023]
Abstract
BACKGROUND There is an increased risk of venous thromboembolism among patients with COVID-19 infection, with the risk being higher among those needing intensive level of care. Existing data is, however limited regarding the outcomes of patients admitted with concurrent COVID-19 infection and pulmonary embolism (PE). METHODS All acute PE admissions were identified from the National Inpatient Sample database during 2020 using ICD-10 codes. Patients were subsequently classified into those with and without COVID-19 infection. The primary outcome of interest was in-hospital mortality. Using multivariate logistic regression, the predictors of mortality were assessed for patients with concurrent acute PE and COVID-19. RESULTS The database query generated 278,840 adult patients with a primary diagnosis of PE. Of these, 4580 patients had concurrent PE and COVID-19 infection. The concurrent PE and COVID-19 infection group had a higher proportion of Black-American and Hispanic patients, and those living in the zip codes associated with lowest annualized income compared to the PE alone group. Furthermore, patients in the concurrent PE and COVID-19 infection group had an increased risk of in-hospital mortality (adjusted odds ratio [aOR]:1.62; 95% CI: 1.17 - 2.24; p = 0.004), septic shock (aOR: 1.66; 95% CI 1.10-2.52; p = 0.016), respiratory failure (aOR: 1.78; 95% CI 1.53-2.06; p = 0.001), and a longer hospital stay [5.5 days vs 4.59 days; p = 0.001). CONCLUSION Concurrent COVID-19 and PE admissions is associated with an increased in-hospital mortality, risk of septic shock and respiratory failure, and a longer length of hospital stay.
Collapse
Affiliation(s)
- Israel Safiriyu
- Department of Medicine, Jacobi Medical Center, Bronx, NY, USA; Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Michael Fatuyi
- Department of Medicine, TriHealth Good Samaritan Hospital Program, Cincinnati Ohio, USA
| | - Adhya Mehta
- Department of Medicine, Jacobi Medical Center, Bronx, NY, USA; Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ahmad Naseer
- Department of Medicine, Jacobi Medical Center, Bronx, NY, USA; Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ebere Alexander
- Department of Medicine, Mt Carmel East Hospital, Colombus, Ohio, USA
| | - Houston Vovan
- Department of Medicine, TriHealth Good Samaritan Hospital Program, Cincinnati Ohio, USA
| | - Garba Rimamskep Shamaki
- Department of Internal medicine Unity Hospital, Rochester Regional Health Rochester, NY, USA
| | - Tamunoinemi Bob-Manuel
- Department of Interventional and Endovascular Cardiology, Stern Cardiovascular Foundation, Memphis, TN, USA
| |
Collapse
|
43
|
Massoud GP, Hazimeh DH, Amin G, Mekary W, Khabsa J, Araji T, Fares S, Mericskay M, Booz GW, Zouein FA. Risk of thromboembolic events in non-hospitalized COVID-19 patients: A systematic review. Eur J Pharmacol 2023; 941:175501. [PMID: 36641102 PMCID: PMC9833853 DOI: 10.1016/j.ejphar.2023.175501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
The risk of thromboembolism in non-hospitalized COVID-19 patients remains uncertain and was assessed in this review to better weigh benefits vs. risks of prophylactic anticoagulation in this population. A search was performed through three databases: Medline, Embase, and Cochrane Library until 2022. Self-controlled case series, case-control and cohort studies were included, and findings summarized narratively. Meta-analyses for risk of thromboembolism including deep vein thrombosis (DVT), pulmonary embolism (PE), and myocardial infarction (MI) between COVID-19 and non-COVID-19 non-hospitalized patients were conducted. Frequency, incidence rate ratio (IRR), and risk ratio (RR) of stroke were used to assess risk in non-hospitalized COVID-19 patients considering the lack of studies to conduct a meta-analysis. Ten studies met inclusion criteria characterized by adult non-hospitalized COVID-19 patients. Risk of bias was relatively low. Risk of DVT (RR: 1.98 with 95% CI: 1.03-3.83) and PE (OR: 6.72 with 95% CI: 4.81-9.39 and RR: 4.44 with 95% CI: 1.98-9.99) increased in non-hospitalized COVID-19 patients compared to controls. Risk of MI (OR: 1.91 with 95% CI: 0.89-4.09) is possibly increased in non-hospitalized COVID-19 patients with moderate certainty when compared to controls. A trend in favor of stroke was documented in the first week following infection. Our meta-analyses support the increase in risk of DVT and PE, and likely increase of MI, in non-hospitalized COVID-19 patients. The risk of stroke appears significant in the first week following infection but drops to insignificance two weeks later. More studies are needed to establish evidence-based recommendations for prophylactic anticoagulation therapy in non-hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Gaelle P Massoud
- Department of Pharmacology and Toxicology, American University of Beirut Medical Center, Faculty of Medicine, Beirut, Lebanon; The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Faculty of Medicine, Beirut, Lebanon; Department of Gynecology and Obstetrics, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - Dana H Hazimeh
- Department of Pharmacology and Toxicology, American University of Beirut Medical Center, Faculty of Medicine, Beirut, Lebanon; The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Faculty of Medicine, Beirut, Lebanon; Department of Gynecology and Obstetrics, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - Ghadir Amin
- Department of Pharmacology and Toxicology, American University of Beirut Medical Center, Faculty of Medicine, Beirut, Lebanon; The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Faculty of Medicine, Beirut, Lebanon
| | - Wissam Mekary
- Department of Pharmacology and Toxicology, American University of Beirut Medical Center, Faculty of Medicine, Beirut, Lebanon; The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Faculty of Medicine, Beirut, Lebanon
| | - Joanne Khabsa
- Clinical Research Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Tarek Araji
- Parker Institute of Cancer and Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Souha Fares
- Hariri School of Nursing, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Center of Innovative Design and Analysis, Biostatistics and Informatics, School of Public Health, Colorado University Anschutz Medical Campus, USA
| | - Mathias Mericskay
- Department of Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Inserm, Université Paris-Saclay, France
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, American University of Beirut Medical Center, Faculty of Medicine, Beirut, Lebanon; The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Faculty of Medicine, Beirut, Lebanon; Department of Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Inserm, Université Paris-Saclay, France; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
44
|
Carnevale R, Cammisotto V, Bartimoccia S, Nocella C, Castellani V, Bufano M, Loffredo L, Sciarretta S, Frati G, Coluccia A, Silvestri R, Ceccarelli G, Oliva A, Venditti M, Pugliese F, Maria Mastroianni C, Turriziani O, Leopizzi M, D'Amati G, Pignatelli P, Violi F. Toll-Like Receptor 4-Dependent Platelet-Related Thrombosis in SARS-CoV-2 Infection. Circ Res 2023; 132:290-305. [PMID: 36636919 DOI: 10.1161/circresaha.122.321541] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND SARS-CoV-2 is associated with an increased risk of venous and arterial thrombosis, but the underlying mechanism is still unclear. METHODS We performed a cross-sectional analysis of platelet function in 25 SARS-CoV-2 and 10 healthy subjects by measuring Nox2 (NADPH oxidase 2)-derived oxidative stress and thromboxane B2, and investigated if administration of monoclonal antibodies against the S protein (Spike protein) of SARS-CoV-2 affects platelet activation. Furthermore, we investigated in vitro if the S protein of SARS-CoV-2 or plasma from SARS-CoV-2 enhanced platelet activation. RESULTS Ex vivo studies showed enhanced platelet Nox2-derived oxidative stress and thromboxane B2 biosynthesis and under laminar flow platelet-dependent thrombus growth in SARS-CoV-2 compared with controls; both effects were lowered by Nox2 and TLR4 (Toll-like receptor 4) inhibitors. Two hours after administration of monoclonal antibodies, a significant inhibition of platelet activation was observed in patients with SARS-CoV-2 compared with untreated ones. In vitro study showed that S protein per se did not elicit platelet activation but amplified the platelet response to subthreshold concentrations of agonists and functionally interacted with platelet TLR4. A docking simulation analysis suggested that TLR4 binds to S protein via three receptor-binding domains; furthermore, immunoprecipitation and immunofluorescence showed S protein-TLR4 colocalization in platelets from SARS-CoV-2. Plasma from patients with SARS-CoV-2 enhanced platelet activation and Nox2-related oxidative stress, an effect blunted by TNF (tumor necrosis factor) α inhibitor; this effect was recapitulated by an in vitro study documenting that TNFα alone promoted platelet activation and amplified the platelet response to S protein via p47phox (phagocyte oxidase) upregulation. CONCLUSIONS The study identifies 2 TLR4-dependent and independent pathways promoting platelet-dependent thrombus growth and suggests inhibition of TLR4. or p47phox as a tool to counteract thrombosis in SARS-CoV-2.
Collapse
Affiliation(s)
- Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (R.C., S.S., G.F., M.L.).,IRCCS Neuromed, Località Camerelle, Pozzilli (IS), Italy (R.C., S.S., G.F.)
| | - Vittoria Cammisotto
- Department of Clinical Internal, Anaesthesiological and Cardiovascular Sciences (V. Cammisotto, S.B., C.N., L.L., P.P.), Sapienza University of Rome, Italy
| | - Simona Bartimoccia
- Department of Clinical Internal, Anaesthesiological and Cardiovascular Sciences (V. Cammisotto, S.B., C.N., L.L., P.P.), Sapienza University of Rome, Italy
| | - Cristina Nocella
- Department of Clinical Internal, Anaesthesiological and Cardiovascular Sciences (V. Cammisotto, S.B., C.N., L.L., P.P.), Sapienza University of Rome, Italy
| | - Valentina Castellani
- Department of General Surgery and Surgical Speciality (V. Castellani, F.P.), Sapienza University of Rome, Italy
| | - Marianna Bufano
- Laboratory affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies (M.B., A.C., R.S.), Sapienza University of Rome, Italy
| | - Lorenzo Loffredo
- Department of Clinical Internal, Anaesthesiological and Cardiovascular Sciences (V. Cammisotto, S.B., C.N., L.L., P.P.), Sapienza University of Rome, Italy
| | - Sebastiano Sciarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (R.C., S.S., G.F., M.L.).,IRCCS Neuromed, Località Camerelle, Pozzilli (IS), Italy (R.C., S.S., G.F.)
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (R.C., S.S., G.F., M.L.).,IRCCS Neuromed, Località Camerelle, Pozzilli (IS), Italy (R.C., S.S., G.F.)
| | - Antonio Coluccia
- Laboratory affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies (M.B., A.C., R.S.), Sapienza University of Rome, Italy
| | - Romano Silvestri
- Laboratory affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies (M.B., A.C., R.S.), Sapienza University of Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases (G.C., A.O., M.V., C.M.M.), Sapienza University of Rome, Italy
| | - Alessandra Oliva
- Department of Public Health and Infectious Diseases (G.C., A.O., M.V., C.M.M.), Sapienza University of Rome, Italy
| | - Mario Venditti
- Department of Public Health and Infectious Diseases (G.C., A.O., M.V., C.M.M.), Sapienza University of Rome, Italy
| | - Francesco Pugliese
- Department of General Surgery and Surgical Speciality (V. Castellani, F.P.), Sapienza University of Rome, Italy
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases (G.C., A.O., M.V., C.M.M.), Sapienza University of Rome, Italy
| | - Ombretta Turriziani
- Laboratory of Virology, Department of Molecular Medicine (O.T.), Sapienza University of Rome, Italy
| | - Martina Leopizzi
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (R.C., S.S., G.F., M.L.)
| | - Giulia D'Amati
- Department of Radiological, Oncological and Pathological Sciences (G.D.), Sapienza University of Rome, Italy
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anaesthesiological and Cardiovascular Sciences (V. Cammisotto, S.B., C.N., L.L., P.P.), Sapienza University of Rome, Italy.,Mediterranea Cardiocentro- Napoli, Italy (P.P., F.V.)
| | | |
Collapse
|
45
|
Candeloro M, Schulman S. Arterial Thrombotic Events in Hospitalized COVID-19 Patients: A Short Review and Meta-Analysis. Semin Thromb Hemost 2023; 49:47-54. [PMID: 35793687 DOI: 10.1055/s-0042-1749661] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It is well established that the risk of venous thromboembolism is high in coronavirus disease 19 (COVID-19). The frequency of arterial thromboembolic events (ATEs) in hospitalized patients with COVID-19 is unclear, as is the magnitude of these events in comparison with other infections. We searched MEDLINE from February 2020 to February 2022 for prospective or retrospective cohort studies and randomized clinical trials that reported the number of acute myocardial infarction (AMI), acute ischemic stroke (AIS), acute limb ischemia (ALI), or other ATE as defined by the original authors in hospitalized patients with COVID-19. The pooled frequencies were calculated through meta-analysis using random effects model with logit transformation and presented with relative 95% prediction intervals (95% PI). We retrieved a total of 4,547 studies, 36 of which (28 retrospective cohorts, five prospective cohorts and three randomized trials) were finally included in our analysis. The resulting cohort counted 100,949 patients, 2,641 (2.6%) of whom experienced ATE. The pooled ATE frequency was 2.0% (95% PI, 0.4-9.6%). The pooled ATE frequency for AMI, AIS, ALI, and other ATE was 0.8% (95% PI, 0.1-8.1%), 0.9% (95% PI, 0.3-2.9%), 0.2% (95% PI, 0.0-4.2%), and 0.5% (95% PI, 0.1-3.0%), respectively. In comparison with the ATE incidence reported in three studies on non-COVID viral pneumonia, we did not detect a significant difference from the results in our analysis. In conclusion, we found a non-negligible proportion of ATE in patients hospitalized for COVID-19. Our results are similar to those found in hospitalized patients with influenza or with non-COVID viral pneumonia.
Collapse
Affiliation(s)
- Matteo Candeloro
- Department of Innovative Technologies in Medicine and Dentistry, "G. D'Annunzio" University, Chieti, Italy.,Department of Medicine, Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Sam Schulman
- Department of Medicine, Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Obstetrics and Gynecology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
46
|
Buso G, Mazzolai L, Rueda-Camino JA, Fernández-Capitán C, Jiménez D, Bikdeli B, Lobo JL, Fernández-Reyes JL, Ciammaichella M, Monreal M. Pulmonary Embolism in Patients with COVID-19: Comparison between Different Care Settings. Semin Thromb Hemost 2023; 49:34-46. [PMID: 34902865 DOI: 10.1055/s-0041-1740152] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The clinical characteristics and outcomes of patients with coronavirus disease 2019 (COVID-19) who develop pulmonary embolism (PE) in the full spectrum of patient care settings need to be elucidated. The aim of this study was to compare the clinical characteristics, treatment, and 90-day outcomes in patients diagnosed with PE while recovering from COVID-19 in the outpatient setting versus those who were diagnosed with PE while being hospitalized with COVID-19. Data from the international Registro Informatizado de Enfermedad TromboEmbólica (RIETE) registry were used. The major study outcomes were all-cause death, major bleeding, and venous thromboembolism (VTE) recurrences during the first 90 days after PE. From March 2020 to March 2021, 737 patients with COVID-19 experienced acute PE. Of these, 340 (46%) were recovering from COVID-19 as outpatients (267 patients who had been treated at home for COVID-19 and 73 discharged after being hospitalized with COVID-19). Compared with inpatients with COVID-19, those recovering in the outpatient setting upon PE were less likely to be men (odds ratio [OR]: 0.54; 95% confidence interval [CI]: 0.40-0.72) and less likely to have hypertension (OR: 0.55; 95% CI: 0.41-0.74) or diabetes (OR: 0.51; 95% CI: 0.33-0.76). At 90-day follow-up, eight patients (none recovering from COVID-19 as outpatient vs. 2.4% of inpatients with COVID-19) developed recurrent VTE, 34 (1.9 vs. 7.9%) had major bleeding, and 128 (10 vs. 24%) died. On multivariable analysis, inpatients with COVID-19 were at a higher risk of major bleeding (adjusted hazard ratio [HR]: 6.80; 95% CI: 1.52-30.4) or death (adjusted HR: 2.24; 95% CI: 1.40-3.58). In conclusion, using a large multinational registry of patients with COVID-19 who experienced PE, thromboembolic episodes occurring in those recovering from COVID-19 as outpatients were associated with less ominous outcomes than inpatients with COVID-19.
Collapse
Affiliation(s)
- Giacomo Buso
- Angiology Division, Heart and Vessels Department, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Lucia Mazzolai
- Angiology Division, Heart and Vessels Department, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | | | | | - David Jiménez
- Respiratory Department, Hospital Ramón y Cajal (IRYCIS), Madrid, Spain.,Medicine Department, Universidad de Alcala, Madrid, Spain
| | - Behnood Bikdeli
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Yale/YNHH Center for Outcomes Research & Evaluation (CORE), New Haven, Connecticut.,Cardiovascular Research Foundation (CRF), New York, New York
| | - José Luis Lobo
- Department of Pneumonology, Hospital Universitario Araba, Álava, Spain
| | | | | | - Manuel Monreal
- Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol. Badalona, Barcelona, Spain.,Chair for the Study of Thromboembolic Disease, Faculty of Health Sciences, UCAM - Universidad Católica San Antonio de Murcia, Spain
| | | |
Collapse
|
47
|
Rizk JG, Gupta A, Lazo JG, Sardar P, Henry BM, Lavie CJ, Effron MB. To Anticoagulate or Not to Anticoagulate in COVID-19: Lessons after 2 Years. Semin Thromb Hemost 2023; 49:62-72. [PMID: 35468641 DOI: 10.1055/s-0042-1744302] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A hypercoagulable state associated with coronavirus disease 2019 (COVID-19) has been well documented and is believed to be strongly supported by a proinflammatory state. The hypercoagulable state in turn results in increased incidence of arterial and venous thromboembolism (VTE) seen in hospitalized COVID-19 when compared with hospitalized non-COVID-19 patient cohorts. Moreover, patients with arterial or VTE and COVID-19 have higher mortality compared with COVID-19 patients without arterial or VTE. Prevention of arterial or VTE thus remains an essential question in the management of COVID-19 patients, especially because of high rates of reported microvascular and macrovascular thrombosis. This has prompted multiple randomized control trials (RCTs) evaluating different anticoagulation strategies in COVID-19 patients at various stages of the disease. Herein, we review findings from RCTs in the past 2 years of antithrombotic therapy in critically ill hospitalized patients, noncritically ill hospitalized patients, patients postdischarge from the hospital, and outpatients. RCTs in critically ill patients demonstrated therapeutic dose anticoagulation does not improve outcomes and has more bleeding than prophylaxis dose anticoagulant in these patients. Trials in noncritically ill hospitalized patients showed a therapeutic dose anticoagulation with a heparin formulation might improve clinical outcomes. Anticoagulation with a direct oral anticoagulant posthospital discharge may improve outcomes, although there is a large RCT in progress. Nonhospitalized COVID-19 patients have an insufficient burden of events to be candidates for antithrombotic therapy. Anticoagulation in pregnant and lactating patients with COVID-19, as well as antiplatelet therapy for COVID-19, is also reviewed.
Collapse
Affiliation(s)
- John G Rizk
- Department of Pharmaceutical Health Services Research, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Aashish Gupta
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-The University of Queensland School of Medicine, New Orleans, Louisiana
| | - Jose G Lazo
- UCSF Medical Center, University of California, San Francisco, San Francisco, California
| | - Partha Sardar
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-The University of Queensland School of Medicine, New Orleans, Louisiana
| | - Brandon Michael Henry
- Cardiac Intensive Care Unit, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Disease Prevention and Intervention & Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-The University of Queensland School of Medicine, New Orleans, Louisiana
| | - Mark B Effron
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-The University of Queensland School of Medicine, New Orleans, Louisiana
| |
Collapse
|
48
|
Abstract
Shortly after the emergence of coronavirus disease 2019 (COVID-19) in late 2019, clinicians rapidly recognized an apparent association between the disease and both arterial and venous thrombotic complications, which was confirmed in epidemiologic studies. Based on these data, hospitals empirically developed and implemented protocols with different strategies for anticoagulation of hospitalized COVID-19 patients. Subsequent randomized controlled trials (RCTs) clarified the role of anticoagulation in patients hospitalized with COVID-19 and recently discharged from the hospital. In this review, we discuss the epidemiology and pathophysiology of thrombosis in patients with COVID-19, observational comparative effectiveness analyses that provided hints of a benefit from anticoagulation, and finally the RCTs that established which patients with COVID-19 benefit from treatment-dose anticoagulation. These RCTs have demonstrated that hospitalized, noncritically ill patients with COVID-19 benefit from treatment-dose anticoagulation, but patients who are hospitalized and critically ill, discharged from the hospital, or not hospitalized do not benefit.
Collapse
Affiliation(s)
- Alexander C Fanaroff
- Penn Cardiovascular Outcomes, Quality and Evaluative Research Center, University of Pennsylvania; Leonard Davis Institute, University of Pennsylvania; Cardiovascular Medicine Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Renato D Lopes
- Division of Cardiovascular Medicine and Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA;
| |
Collapse
|
49
|
Gjonbalaj N, Uka S, Olluri E, Sulovari A, Vishaj M, Kamberi L, Berisha H, Gjonbalaj E. Renal artery thrombosis as a long-term complication of COVID-19. Radiol Case Rep 2023; 18:260-265. [PMID: 36345461 PMCID: PMC9631237 DOI: 10.1016/j.radcr.2022.10.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/03/2022] [Accepted: 10/08/2022] [Indexed: 11/05/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been extensively associated with microvascular and macrovascular thrombosis. Several reports have demonstrated a link between COVID-19 and pulmonary embolism, deep vein thrombosis, myocardial infarction, stroke, and aortic thrombosis. Renal artery thrombosis is of special interest because of its life-threatening consequences, such as acute kidney injury and renal infarction. We present a case of left renal artery thrombosis as a long-term complication of COVID-19. Moreover, we demonstrate the effectiveness of interventional radiology to regain vascularization of the affected kidney.
Collapse
Affiliation(s)
- Naser Gjonbalaj
- Radiology Clinic, University Clinical Center of Kosovo, Prishtina 10000, Republic of Kosovo, XK
- Department of Radiology, American Hospital of Kosovo, Rruga Shkupi, Nr. 25, Pristina 10000, Republic of Kosovo, XK
- Alma Mater Europaea Campus Colege Rezonanca, Hospital district Prishtine, 10000, Republic of Kosovo, XK
| | - Safet Uka
- Radiology Clinic, University Clinical Center of Kosovo, Prishtina 10000, Republic of Kosovo, XK
| | - Elmi Olluri
- Vascular Surgery Clinic, University Clinical Center of Kosovo, Prishtina 10000, Republic of Kosovo, XK
- Department of Vascular Surgery, American Hospital of Kosovo, Rruga Shkupi, Nr. 25, Pristina 10000, Republic of Kosovo, XK
| | - Admir Sulovari
- Department of Radiology, American Hospital of Kosovo, Rruga Shkupi, Nr. 25, Pristina 10000, Republic of Kosovo, XK
| | - Murat Vishaj
- Department of Anesthesiology, American Hospital of Kosovo, Rruga Shkupi, Nr. 25, Pristina 10000, Republic of Kosovo, XK
| | - Lendita Kamberi
- Department of Radiology, American Hospital of Kosovo, Rruga Shkupi, Nr. 25, Pristina 10000, Republic of Kosovo, XK
| | - Hamez Berisha
- Department of Emergency, American Hospital of Kosovo, Rruga Shkupi, Nr. 25, Pristina 10000, Republic of Kosovo, XK
| | - Enkelejda Gjonbalaj
- Department of Pharmacy, University Clinical Center of Kosovo, Prishtina 10000, Republic of Kosovo, XK
| |
Collapse
|
50
|
Sensen B, Wichmann D, Kluge S. [Drug therapy interventions for acute SARS-CoV-2 infection: who, when, and what?]. Pneumologie 2023; 77:21-26. [PMID: 36691378 DOI: 10.1055/a-1983-3800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
There is a variety of drug therapy options for treatment of acute SARS-CoV-2 infections. The updated S3 guideline "Recommendations for inpatient therapy of patients with COVID-19" provides clear recommendations in this regard. Which therapy is best suited for which patient and in which phase of the disease must be decided individually based on risk factors, comorbidities and contraindications. This article provides an overview.
Collapse
Affiliation(s)
- Barbara Sensen
- Klinik für Intensivmedizin des Zentrums für Anästhesiologie und Intensivmedizin am Universitätsklinikum, Hamburg-Eppendorf, Hamburg
| | - Dominic Wichmann
- Klinik für Intensivmedizin des Zentrums für Anästhesiologie und Intensivmedizin am Universitätsklinikum, Hamburg-Eppendorf, Hamburg
| | - Stefan Kluge
- Klinik für Intensivmedizin des Zentrums für Anästhesiologie und Intensivmedizin am Universitätsklinikum, Hamburg-Eppendorf, Hamburg
| |
Collapse
|