101
|
Kim IS, Lee SG, Shin SG, Jeong H, Sohn KM, Park KS, Silwal P, Cheon S, Kim J, Kym S, Kim YS, Jo EK, Park C. Dysregulated thrombospondin 1 and miRNA-29a-3p in severe COVID-19. Sci Rep 2022; 12:21227. [PMID: 36481664 PMCID: PMC9732043 DOI: 10.1038/s41598-022-23533-x] [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: 05/05/2022] [Accepted: 10/31/2022] [Indexed: 12/13/2022] Open
Abstract
Although nearly a fifth of symptomatic COVID-19 patients suffers from severe pulmonary inflammation, the mechanism of developing severe illness is not yet fully understood. To identify significantly altered genes in severe COVID-19, we generated messenger RNA and micro-RNA profiling data of peripheral blood mononuclear cells (PBMCs) from five COVID-19 patients (2 severe and 3 mild patients) and three healthy controls (HC). For further evaluation, two publicly available RNA-Seq datasets (GSE157103 and GSE152418) and one single-cell RNA-Seq dataset (GSE174072) were employed. Based on RNA-Seq datasets, thrombospondin 1 (THBS1) and interleukin-17 receptor A (IL17RA) were significantly upregulated in severe COVID-19 patients' blood. From single-cell RNA-sequencing data, IL17RA level is increased in monocytes and neutrophils, whereas THBS1 level is mainly increased in the platelets. Moreover, we identified three differentially expressed microRNAs in severe COVID-19 using micro-RNA sequencings. Intriguingly, hsa-miR-29a-3p significantly downregulated in severe COVID-19 was predicted to bind the 3'-untranslated regions of both IL17RA and THBS1 mRNAs. Further validation analysis of our cohort (8 HC, 7 severe and 8 mild patients) showed that THBS1, but not IL17RA, was significantly upregulated, whereas hsa-miR-29a-3p was downregulated, in PBMCs from severe patients. These findings strongly suggest that dysregulated expression of THBS1, IL17RA, and hsa-miR-29a-3p involves severe COVID-19.
Collapse
Affiliation(s)
- In Soo Kim
- grid.254230.20000 0001 0722 6377Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Sung-Gwon Lee
- grid.14005.300000 0001 0356 9399School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Seul Gi Shin
- grid.254230.20000 0001 0722 6377Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hyeongseok Jeong
- grid.254230.20000 0001 0722 6377Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Kyung Mok Sohn
- grid.254230.20000 0001 0722 6377Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Ki-Sun Park
- grid.418980.c0000 0000 8749 5149KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - Prashanta Silwal
- grid.254230.20000 0001 0722 6377Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Shinhye Cheon
- grid.254230.20000 0001 0722 6377Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jungok Kim
- grid.254230.20000 0001 0722 6377Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Sungmin Kym
- grid.254230.20000 0001 0722 6377Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Yeon-Sook Kim
- grid.254230.20000 0001 0722 6377Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Eun-Kyeong Jo
- grid.254230.20000 0001 0722 6377Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Chungoo Park
- grid.14005.300000 0001 0356 9399School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| |
Collapse
|
102
|
Huang RC, Chiu CH, Shang HS, Perng CL, Chiang TT, Tsai CC, Wang CH. Clinical characteristics analysis of COVID-19 patients from the first significant community outbreak by SARS-CoV-2 variant B.1.1.7 in Taiwan as experienced from a single northern medical center. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2022; 55:1036-1043. [PMID: 36057491 PMCID: PMC9381423 DOI: 10.1016/j.jmii.2022.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 07/11/2022] [Accepted: 08/01/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND/PURPOSE Clinical characteristics of patients in the first community outbreak of coronavirus disease 2019 (COVID-19) by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant B.1.1.7 in Taiwan have not been characterized. METHODS SARS-CoV-2 positive specimens from inpatients between May 7 and June 15 in 2021were screen for SARS-CoV-2 B.1.1.7 lineage by VirSNiP assay. Clinical characteristics were reviewed and compared with those from Feb 1 to April 30, 2020 and from Jan 1 to March 31, 2022. RESULTS One hundred forty-one inpatients from May 7 to June 15, 2021 infected with SARS-CoV-2 B.1.1.7 lineage were included. The major presenting symptoms were fever (88.7%) and cough (59.6%). Incidence of relevant complications including pulmonary embolism, simultaneous infections with bacteria, virus, and fungi were 0.7%, 12.8%, 13.5%, and 2.1%, respectively. Old age, high Charlson comorbidity index, short of breath, and initial critical illness were independently associated with 28-day mortality (all p < 0.05). In comparison to COVID-19 inpatients from Feb 1 to April 30, 2020, patients from the outbreak by SARS-CoV-2 B.1.1.7 lineage were older, more severe in disease condition, higher mortality but less obvious initial presenting symptoms. After implementation of nationwide vaccination campaign in the next half year of 2021, COVID-19 inpatients from Jan 1 to March 31 in 2022 indicated less severe diseases than those infected with SARS-CoV-2 B.1.1.7 lineage. CONCLUSION COVID-19 inpatients by SARS-CoV-2 variant B.1.1.7 with old age, multiple comorbidities, and more severe disease conditions were associated with increased mortality. Vaccination for this vulnerable populations may be helpful.
Collapse
Affiliation(s)
- Ruei-Chang Huang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chun-Hsiang Chiu
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hung-Sheng Shang
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Cherng-Lih Perng
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Tsung-Ta Chiang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chun-Chou Tsai
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Ching-Hsun Wang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC,Corresponding author. Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, No. 325, Section 2, Cheng-Kung Road, Neihu 114, Taipei, Taiwan. Fax: +886-2-87927258
| |
Collapse
|
103
|
Urano T, Yasumoto A, Yokoyama K, Horiuchi H, Morishita E, Suzuki Y. COVID-19 and Thrombosis: Clinical Aspects. Curr Drug Targets 2022; 23:1567-1572. [PMID: 36200150 DOI: 10.2174/1389450123666221005092350] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/30/2022] [Accepted: 09/09/2022] [Indexed: 01/25/2023]
Abstract
In coronavirus disease 2019 (COVID-19), thrombus formation is related to the pathogenesis of acute respiratory distress syndrome (ARDS) and the progression of clinical symptoms. Severe damage to vascular endothelial cells and the associated cytokine storm after SARS-CoV-2 infection cause thrombogenesis and contribute to the development of more severe and unique thromboses compared to other infectious diseases. Thromboses occur more often in critically ill patients. In addition to pulmonary thromboembolism (PE) and deep vein thrombosis, acute myocardial infarction, peripheral arterial thrombosis, and aortic thrombosis have also been reported. In PE, thrombi develop in both pulmonary arteries and alveolar capillaries. These, together with intraalveolar fibrin deposition, interfere with effective gaseous exchange in the lungs and exacerbate the clinical symptoms of ARDS in patients with COVID-19. Pharmacological thromboprophylaxis is recommended for all hospitalized patients to prevent both thrombosis and aggravation of ARDS, and other organ failures. Although the pediatric population is mostly asymptomatic or develops mild disease after SARS-CoV-2 infection, a new inflammatory disorder affecting the cardiovascular system, multisystem inflammatory syndrome in children (MIS-C), has been reported. Similar to Kawasaki disease, acute myocarditis, coronary vasculitis, and aneurysms are typically seen in MISC, although these two are now considered distinct entities. A similar acute myocarditis is also observed in young male adults, in which a hyperinflammatory state after SARS-CoV-2 infection seems to be involved. Several side effects following vaccination against COVID-19 have been reported, including vaccine-induced immune thrombotic thrombocytopenia and acute myocarditis. Although these could be serious and life-threatening, the cases are very rare, thus, the benefits of immunization still outweigh the risks.
Collapse
Affiliation(s)
- Tetsumei Urano
- Department of Medical Physiology, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Atsushi Yasumoto
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sappero, Japan
| | - Kenji Yokoyama
- Department of Hematology/Oncology, Tokai University, Tokyo, Japan
| | - Hisanori Horiuchi
- Department of Molecular and Cellular Biology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Eriko Morishita
- Department of Clinical Laboratory Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yuko Suzuki
- Department of Medical Physiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
104
|
Kangro K, Wolberg AS, Flick MJ. Fibrinogen, Fibrin, and Fibrin Degradation Products in COVID-19. Curr Drug Targets 2022; 23:1593-1602. [PMID: 36029073 DOI: 10.2174/1389450123666220826162900] [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: 03/29/2022] [Revised: 05/31/2022] [Accepted: 06/15/2022] [Indexed: 01/25/2023]
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is the highly pathogenic and highly transmissible human coronavirus that is the causative agent for the worldwide COVID-19 pandemic. COVID-19 manifests predominantly as a respiratory illness with symptoms consistent with viral pneumonia, but other organ systems (e.g., kidney, heart, brain) can also become perturbed in COVID-19 patients. Accumulating data suggest that significant activation of the hemostatic system is a common pathological manifestation of SARS-CoV-2 infection. The clotting protein fibrinogen is one of the most abundant plasma proteins. Following activation of coagulation, the central coagulation protease thrombin converts fibrinogen to fibrin monomers, which selfassemble to form a matrix, the primary structural component of the blood clot. Severe COVID-19 is associated with a profound perturbation of circulating fibrinogen, intra- and extravascular fibrin deposition and persistence, and fibrin degradation. Current findings suggest high levels of fibrinogen and the fibrin degradation product D-dimer are biomarkers of poor prognosis in COVID-19. Moreover, emerging studies with in vitro and animal models indicate fibrin(ogen) as an active player in COVID-19 pathogenesis. Here, we review the current literature regarding fibrin(ogen) and COVID-19, including possible pathogenic mechanisms and treatment strategies centered on clotting and fibrin(ogen) function.
Collapse
Affiliation(s)
- Kadri Kangro
- Department of Pathology and Laboratory Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew J Flick
- Department of Pathology and Laboratory Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
105
|
Jing H, Wu X, Xiang M, Liu L, Novakovic VA, Shi J. Pathophysiological mechanisms of thrombosis in acute and long COVID-19. Front Immunol 2022; 13:992384. [PMID: 36466841 PMCID: PMC9709252 DOI: 10.3389/fimmu.2022.992384] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/27/2022] [Indexed: 08/02/2023] Open
Abstract
COVID-19 patients have a high incidence of thrombosis, and thromboembolic complications are associated with severe COVID-19 and high mortality. COVID-19 disease is associated with a hyper-inflammatory response (cytokine storm) mediated by the immune system. However, the role of the inflammatory response in thrombosis remains incompletely understood. In this review, we investigate the crosstalk between inflammation and thrombosis in the context of COVID-19, focusing on the contributions of inflammation to the pathogenesis of thrombosis, and propose combined use of anti-inflammatory and anticoagulant therapeutics. Under inflammatory conditions, the interactions between neutrophils and platelets, platelet activation, monocyte tissue factor expression, microparticle release, and phosphatidylserine (PS) externalization as well as complement activation are collectively involved in immune-thrombosis. Inflammation results in the activation and apoptosis of blood cells, leading to microparticle release and PS externalization on blood cells and microparticles, which significantly enhances the catalytic efficiency of the tenase and prothrombinase complexes, and promotes thrombin-mediated fibrin generation and local blood clot formation. Given the risk of thrombosis in the COVID-19, the importance of antithrombotic therapies has been generally recognized, but certain deficiencies and treatment gaps in remain. Antiplatelet drugs are not in combination with anticoagulant treatments, thus fail to dampen platelet procoagulant activity. Current treatments also do not propose an optimal time for anticoagulation. The efficacy of anticoagulant treatments depends on the time of therapy initiation. The best time for antithrombotic therapy is as early as possible after diagnosis, ideally in the early stage of the disease. We also elaborate on the possible mechanisms of long COVID thromboembolic complications, including persistent inflammation, endothelial injury and dysfunction, and coagulation abnormalities. The above-mentioned contents provide therapeutic strategies for COVID-19 patients and further improve patient outcomes.
Collapse
Affiliation(s)
- Haijiao Jing
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
| | - Xiaoming Wu
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
| | - Mengqi Xiang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
| | - Langjiao Liu
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
| | - Valerie A. Novakovic
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA, United States
| | - Jialan Shi
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
106
|
Leither LM, Buckel W, Brown SM. Care of the Seriously Ill Patient with SARS-CoV-2. Med Clin North Am 2022; 106:949-960. [PMID: 36280338 PMCID: PMC9364720 DOI: 10.1016/j.mcna.2022.08.002] [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] [Indexed: 12/15/2022]
Abstract
In late 2019, SARS-CoV-2 caused the greatest global health crisis in a century, impacting all aspects of society. As the COVID-19 pandemic evolved throughout 2020 and 2021, multiple variants emerged, contributing to multiple surges in cases of COVID-19 worldwide. In 2021, highly effective vaccines became available, although the pandemic continues into 2022. There has been tremendous expansion of basic, translational, and clinical knowledge about SARS-CoV-2 and COVID-19 since the pandemic's onset. Treatment options have been rapidly explored, attempting to repurpose preexisting medications in tandem with development and evaluation of novel agents. Care of the seriously ill patient is examined.
Collapse
Affiliation(s)
- Lindsay M Leither
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Intermountain Medical Center, 5121 South Cottonwood Street, Salt Lake City, UT 84107, USA; Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA.
| | - Whitney Buckel
- Pharmacy Services, Intermountain Healthcare, 4393 S Riverboat Road, Taylorsville, UT 84123, USA
| | - Samuel M Brown
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Intermountain Medical Center, 5121 South Cottonwood Street, Salt Lake City, UT 84107, USA; Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
107
|
Inflammatory and Hemostatic Markers in COVID-19 Patients with Arterial Thrombosis Are Significantly Lower at Hospital Admission than in COVID-19 Patients without Thrombosis. Viruses 2022; 14:v14112330. [PMID: 36366428 PMCID: PMC9693543 DOI: 10.3390/v14112330] [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: 09/22/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 02/01/2023] Open
Abstract
Patients with Coronavirus disease 2019 (COVID-19) are at increased risk of venous thromboembolism (VTE); however, data on arterial thromboembolism (ATE) is still limited. We report a case series of thromboembolic events (TE) in 290 COVID-19 patients admitted between October and December 2020 to a Portuguese hospital. Admission levels of various laboratory parameters were evaluated and compared between COVID-19 patients with (TE) and without thrombotic events (non-TE). The overall incidence of isolated ATE was 5.52%, isolated VTE was 2.41% and multiple mixed events was 0.7%. A total of 68% events were detected upon admission to the hospital with 76% corresponding to ATE. Admissions to the Intensive Care Unit were higher in patients with TE, when comparing with the non-TE group (44% vs. 27.2%; p = 0.003). Patients with ATE presented significantly lower levels of CRP (p = 0.007), ferritin (p = 0.045), LDH (p = 0.037), fibrinogen (p = 0.010) and higher monocyte counts (p = 0.033) comparatively to the non-TE patients. These results point to an early occurrence of TE and an increased incidence of ATE over VTE. The less prominent inflammation markers in patients with TE and the early presence of TE in patients with otherwise no reason for hospitalization, may suggest a direct role of SARS-CoV-2 in the thrombotic process.
Collapse
|
108
|
Prameswari HS, Putra ICS, Raffaello WM, Nathaniel M, Suhendro AS, Khalid AF, Pranata R. Managing Covid-19 in patients with heart failure: current status and future prospects. Expert Rev Cardiovasc Ther 2022; 20:807-828. [PMID: 36185009 DOI: 10.1080/14779072.2022.2132230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
INTRODUCTION COVID-19 may contribute to decompensation of previously stable chronic HF or cause a de-novo heart failure, which may come from the hyperinflammatory response and subsequent increase in metabolic demand. AREAS COVERED Two independent investigators searched MEDLINE (via PubMed), Europe PMC, and ScienceDirect databases with the following search terms: COVID-19, heart failure, COVID-19 drugs, heart failure drugs, and device therapy. All of the included full-text articles were rigorously evaluated by both authors in case there was disagreement about whether research should be included or not. In total, 157 studies were included and underwent extensive reading by the authors. EXPERT OPINION The World Health Organization (WHO) and the National Institute of Health (NIH) have published COVID-19 drug recommendations, although recommendations for HF-specific drug choices in COVID-19 are still lacking. We hope that this review can answer the void of comprehensive research data regarding the management options of HF in the COVID-19 condition so that clinicians can at least choose a more beneficial therapy or avoid combination therapies that have a high burden of side effects on HF; thus, morbidity and mortality in COVID-19 patients with HF may be reduced.
Collapse
Affiliation(s)
- Hawani Sasmaya Prameswari
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Iwan Cahyo Santosa Putra
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | | | - Michael Nathaniel
- School of Medicine and Health Sciences Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Adrian Sebastian Suhendro
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Achmad Fitrah Khalid
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Raymond Pranata
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
109
|
Nishimoto Y, Yachi S, Takeyama M, 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, Yamashita Y. The current status of thrombosis and anticoagulation therapy in patients with COVID-19 in Japan: From the CLOT-COVID study. J Cardiol 2022; 80:285-291. [PMID: 35430141 PMCID: PMC8979768 DOI: 10.1016/j.jjcc.2022.03.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/15/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Data on thrombosis and current real-world management strategies for anticoagulation therapy are scarce but important for understanding current issues and unmet needs of an optimal management of patients with coronavirus disease 2019 (COVID-19). METHOD The CLOT-COVID Study (thrombosis and antiCoaguLatiOn Therapy in patients with COVID-19 in Japan Study: UMIN000045800) was a retrospective, multicenter cohort study enrolling consecutive hospitalized patients with COVID-19 among 16 centers in Japan from April 2021 to September 2021, and we tried to capture the status of the patients in the fourth and fifth waves of the COVID-19 infections in Japan. We enrolled consecutive hospitalized patients who were diagnosed with COVID-19 and had a positive polymerase chain reaction test obtained from the hospital databases. RESULTS Among 2894 patients with COVID-19, 1245 (43%) received pharmacological thromboprophylaxis. The proportion of pharmacological thromboprophylaxis increased according to the severity of the COVID-19 in 9.8% with mild COVID-19, 61% with moderate COVID-19, and 97% with severe COVID-19. The types and doses of anticoagulants varied widely across the participating centers. During the hospitalization, 38 patients (1.3%) and 126 (4.4%) underwent ultrasound examinations for the lower extremities and contrast-enhanced computed tomography examinations, respectively, and 55 (1.9%) developed thrombosis, mostly venous thromboembolism (71%). The incidence of thrombosis increased according to the severity of the COVID-19 in 0.2% with mild COVID-19, 1.4% with moderate COVID-19, and 9.5% with severe COVID-19. Major bleeding occurred in 57 patients (2.0%) and 158 (5.5%) died, and 81% of them were due to respiratory failure from COVID-19 pneumonia. CONCLUSIONS In the present large-scale observational study, pharmacological thromboprophylaxis for hospitalized patients with COVID-19 was common especially in patients with severe COVID-19, and management strategies varied widely across the participating centers. The overall incidence of thrombosis was substantially low with an increased incidence according to the severity of the COVID-19.
Collapse
Affiliation(s)
- Yuji Nishimoto
- Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, 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
| | | | | | | | | | - Satoshi Ikeda
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | | - Hiroya Hayashi
- Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hirono Satokawa
- Fukushima Medical University, School of Medicine, Fukushima, Japan
| | | | - Eriko Iwata
- Nankai Medical Center Japan Community Health Care Organization, Saiki, Japan
| | | | | | - Akane Kondo
- Shikoku Medical Center for Children and Adults, Zentsuji, Japan
| | | | | | | | | | - Makoto Mo
- Yokohama Minami Kyosai Hospital, Yokohama, Japan
| | | |
Collapse
|
110
|
Schulman S, Sholzberg M, Spyropoulos AC, Zarychanski R, Resnick HE, Bradbury CA, Broxmeyer L, Connors JM, Falanga A, Iba T, Kaatz S, Levy JH, Middeldorp S, Minichiello T, Ramacciotti E, Samama CM, Thachil J. ISTH guidelines for antithrombotic treatment in COVID-19. J Thromb Haemost 2022; 20:2214-2225. [PMID: 35906716 PMCID: PMC9349907 DOI: 10.1111/jth.15808] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/17/2022] [Accepted: 07/03/2022] [Indexed: 11/29/2022]
Abstract
Antithrombotic agents reduce risk of thromboembolism in severely ill patients. Patients with coronavirus disease 2019 (COVID-19) may realize additional benefits from heparins. Optimal dosing and timing of these treatments and benefits of other antithrombotic agents remain unclear. In October 2021, ISTH assembled an international panel of content experts, patient representatives, and a methodologist to develop recommendations on anticoagulants and antiplatelet agents for patients with COVID-19 in different clinical settings. We used the American College of Cardiology Foundation/American Heart Association methodology to assess level of evidence (LOE) and class of recommendation (COR). Only recommendations with LOE A or B were included. Panelists agreed on 12 recommendations: three for non-hospitalized, five for non-critically ill hospitalized, three for critically ill hospitalized, and one for post-discharge patients. Two recommendations were based on high-quality evidence, the remainder on moderate-quality evidence. Among non-critically ill patients hospitalized for COVID-19, the panel gave a strong recommendation (a) for use of prophylactic dose of low molecular weight heparin or unfractionated heparin (LMWH/UFH) (COR 1); (b) for select patients in this group, use of therapeutic dose LMWH/UFH in preference to prophylactic dose (COR 1); but (c) against the addition of an antiplatelet agent (COR 3). Weak recommendations favored (a) sulodexide in non-hospitalized patients, (b) adding an antiplatelet agent to prophylactic LMWH/UFH in select critically ill, and (c) prophylactic rivaroxaban for select patients after discharge (all COR 2b). Recommendations in this guideline are based on high-/moderate-quality evidence available through March 2022. Focused updates will incorporate future evidence supporting changes to these recommendations.
Collapse
Affiliation(s)
- 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
| | - Michelle Sholzberg
- Departments of Medicine, and Laboratory Medicine and Pathobiology, St Michael's Hospital, Li Ka Shing Knowledge Institute, University of Toronto, Toronto, Ontario, Canada
| | - Alex C Spyropoulos
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Ryan Zarychanski
- Sections of Hematology/Oncology and Critical Care, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | - Jean Marie Connors
- Division of Hematology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Anna Falanga
- Department of Transfusion Medicine and Hematology, Hospital Papa Giovanni XXIII, Bergamo, Italy
- University of Milan Bicocca, Monza, Italy
| | - Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University, Tokyo, Japan
| | - Scott Kaatz
- Division of Hospital Medicine, Henry Ford Hospital, Detroit, Michigan, USA
| | - Jerrold H Levy
- Departments of Anesthesiology, Critical Care, and Surgery (Cardiothoracic), Duke University School of Medicine, Durham, North Carolina, USA
| | - Saskia Middeldorp
- Department of Internal Medicine and Radboud Institute of Health Sciences, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Tracy Minichiello
- Division of Hematology, San Francisco VA Medical Center, University of California, San Francisco, San Francisco, California, USA
| | - Eduardo Ramacciotti
- Science Valley Research Institute, São Paulo, Brazil
- Hospital e Maternidade Christóvão da Gama, Grupo Leforte, Santo André, São Paulo, Brazil
| | - Charles Marc Samama
- Department of Anaesthesia, Centre-Université de Paris-Cochin Hospital, Intensive Care and Perioperative Medicine GHU AP-HP, Paris, France
| | - Jecko Thachil
- Department of Haematology, Manchester University Hospitals, Manchester, UK
| |
Collapse
|
111
|
Ramakrishnan N, Ramasubban S, Hegde A, Govil D. Approach to Thromboprophylaxis for Prevention of Venous Thromboembolism in COVID-19: Global Updates and Clinical Insights from India. Clin Pract 2022; 12:766-781. [PMID: 36286066 PMCID: PMC9601217 DOI: 10.3390/clinpract12050080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/30/2022] Open
Abstract
Venous thromboembolism (VTE) frequently occurs in patients with coronavirus disease-19 (COVID-19) and is associated with increased mortality. Several global guidelines recommended prophylactic-intensity anticoagulation rather than intermediate-intensity or therapeutic-intensity anticoagulation for patients with COVID-19-related acute or critical illness without suspected or confirmed VTE. Even though standard doses of thromboprophylaxis are received, many cases of thrombotic complications are reported; hence, appropriate and adequate thromboprophylaxis is critical for the prevention of VTE in COVID-19. In spite of an increased prevalence of VTE in Indian patients, sufficient data on patient characteristics, diagnosis, and therapeutic approach for VTE in COVID is lacking. In this article, we review the available global literature (search conducted up to 31 May 2021) and provide clinical insights into our approach towards managing VTE in patients with COVID-19. Furthermore, in this review, we summarize the incidence and risk factors for VTE with emphasis on the thromboprophylaxis approach in hospitalized patients and special populations with COVID-19 and assess clinical implications in the Indian context.
Collapse
Affiliation(s)
- Nagarajan Ramakrishnan
- Department of Critical Care Medicine, Apollo Hospitals, Chennai 600006, Tamil Nadu, India
| | - Suresh Ramasubban
- Department of Critical Care, Apollo Gleneagles Hospital, Kolkata 700054, West Bengal, India
| | - Ashit Hegde
- Department of Critical Care and Medicine, PD Hinduja National Hospital and Medical Research Centre, Mumbai 400016, Maharashtra, India
| | - Deepak Govil
- Institute of Critical Care and Anesthesiology, Medanta The Medicity, Gurgaon 122006, Haryana, India
| |
Collapse
|
112
|
Hong PY, Huang MH, Hu AK, Lai YT, Zeng HQ, Zhang XB. Use of low molecular weight heparin and hemoglobin fall in COVID-19 patients: A STROBE-compliant study. Medicine (Baltimore) 2022; 101:e30367. [PMID: 36123921 PMCID: PMC9477703 DOI: 10.1097/md.0000000000030367] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
In patients with coronavirus disease 2019 (COVID-19), anticoagulation was suggested as a mitigating strategy. However, little research has been conducted on the adverse consequences of anticoagulant medication. This study aimed to investigate the adverse effect of low molecular weight heparin (LMWH) on hemoglobin fall in COVID-19 treatment. The electronic medical records of COVID-19 patients with pneumonia were collected (including clinical characteristics, vaccination status, complete blood count, coagulation profile, inflammatory cytokines, serum biochemical indicators, and computerized tomography imaging score). Whether they received LMWH, patients were divided into the LMWH group and the control group. Count data were represented as frequency distribution, and a 2-tailed test was used to compare the 2 groups. Spearman rank correlation was used to evaluate the interrelation between changes in hemoglobin and LMWH. The confounding factors were excluded by logistic regression analysis. A total of 179 COVID-19 pneumonia patients were enrolled (81 in the LMWH group and 98 in the control group). The change in hemoglobin was -6.0g/L (IQR -10.8 to 1.0) in the LMWH group and -2.0g/L (IQR -7.0 to 4.0) in the control group (P < .001, between-group difference, -5.0 g/L; 95% confidence interval, -7.0 to -3.0, calculated with the use of the Mann-Whitney U test and the Hodges-Lehmann estimate of confidence intervals for pseudo-medians). The results of multivariate regression analysis showed that after adjusting for confounding factors, LMWH use was not associated with a decrease in hemoglobin (P > .05). In nonsevere COVID-19 patients with pneumonia, the preventive use of LMWH did not lower hemoglobin.
Collapse
Affiliation(s)
- Ping-Yang Hong
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University; the Third Clinical College of Fujian Medical University, Fujian, China
| | - Mao-Hong Huang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University; the Third Clinical College of Fujian Medical University, Fujian, China
| | - An-Ke Hu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University; the Third Clinical College of Fujian Medical University, Fujian, China
| | - Yan-Ting Lai
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University; the Third Clinical College of Fujian Medical University, Fujian, China
| | - Hui-Qing Zeng
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University; the Third Clinical College of Fujian Medical University, Fujian, China
| | - Xiao-Bin Zhang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University; the Third Clinical College of Fujian Medical University, Fujian, China
- *Correspondence: Xiao-Bin Zhang, No. 201, Hubin Nan Road, Siming District, Xiamen, Fujian Province, China (e-mail: )
| |
Collapse
|
113
|
Murakami Y, Okazaki S, Yamamoto M, Sakurai R, Jinno J, Ozono T, Ikenaka K, Gon Y, Todo K, Sasaki T, Hirata H, Uchiyama A, Mochizuki H. Ischemic Stroke Due to Heparin-induced Thrombocytopenia during Severe COVID-19 Infection. Intern Med 2022; 61:2797-2801. [PMID: 35793954 PMCID: PMC9556234 DOI: 10.2169/internalmedicine.9531-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
A 53-year-old woman with severe coronavirus disease 2019 (COVID-19) pneumonia was admitted and treated with intravenous unfractionated heparin for thromboprophylaxis under general anesthesia with mechanical ventilation. She developed right hemiparesis after hospitalization due to a large hemorrhagic infarction. Her platelet count decreased from 243,000/μL at administration to 121,000/μL. Anti-platelet factor 4-heparin antibody testing was positive according to a latex immunoturbidimetric assay. She was therefore diagnosed with heparin-induced thrombocytopenia. We immediately stopped the heparin and started argatroban; the platelet count recovered, and thrombosis did not relapse. Physicians should consider heparin-induced thrombocytopenia as a cause of ischemic stroke in patients with COVID-19 infection.
Collapse
Affiliation(s)
- Yasutaka Murakami
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Shuhei Okazaki
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Makoto Yamamoto
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Japan
| | - Rei Sakurai
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Jyunki Jinno
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Tatsuhiko Ozono
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Kensuke Ikenaka
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Yasufumi Gon
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Kenichi Todo
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Tsutomu Sasaki
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Japan
| | - Akinori Uchiyama
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| |
Collapse
|
114
|
Cuker A, Tseng EK, Nieuwlaat R, Angchaisuksiri P, Blair C, Dane K, DeSancho MT, Diuguid D, Griffin DO, Kahn SR, Klok FA, Lee AI, Neumann I, Pai A, Righini M, Sanfilippo KM, Siegal DM, Skara M, Terrell DR, Touri K, Akl EA, Al Jabiri R, Al Jabiri Y, Barbara AM, Bognanni A, Boulos M, Brignardello-Petersen R, Charide R, Colunga-Lozano LE, Dearness K, Darzi AJ, Hussein H, Karam SG, Mansour R, Morgano GP, Morsi RZ, Muti-Schünemann G, Nadim MK, Philip BA, Qiu Y, Benitez YR, Stevens A, Solo K, Wiercioch W, Mustafa RA, Schünemann HJ. American Society of Hematology living guidelines on the use of anticoagulation for thromboprophylaxis in patients with COVID-19: January 2022 update on the use of therapeutic-intensity anticoagulation in acutely ill patients. Blood Adv 2022; 6:4915-4923. [PMID: 35503027 PMCID: PMC9068240 DOI: 10.1182/bloodadvances.2022007561] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/18/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND COVID-19-related acute illness is associated with an increased risk of venous thromboembolism (VTE). OBJECTIVE These evidence-based guidelines from the American Society of Hematology (ASH) are intended to support patients, clinicians, and other health care professionals in making decisions about the use of anticoagulation in patients with COVID-19. METHODS ASH formed a multidisciplinary guideline panel that included patient representatives and applied strategies to minimize potential bias from conflicts of interest. The McMaster University GRADE Centre supported the guideline development process and performed systematic evidence reviews (through November 2021). The panel prioritized clinical questions and outcomes according to their importance for clinicians and patients. The panel used the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach to assess evidence and make recommendations, which were subject to public comment. This is an update to guidelines published in February 2021 as part of the living phase of these guidelines. RESULTS The panel made one additional recommendation. The panel issued a conditional recommendation in favor of therapeutic-intensity over prophylactic-intensity anticoagulation in patients with COVID-19-related acute illness who do not have suspected or confirmed VTE. The panel emphasized the need for an individualized assessment of risk of thrombosis and bleeding. The panel also noted that heparin (unfractionated or low molecular weight) may be preferred because of a preponderance of evidence with this class of anticoagulants. CONCLUSION This conditional recommendation was based on very low certainty in the evidence, underscoring the need for additional, high-quality, randomized controlled trials comparing different intensities of anticoagulation in patients with COVID-19-related acute illness.
Collapse
Affiliation(s)
- Adam Cuker
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eric K. Tseng
- Division of Hematology/Oncology, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| | - Robby Nieuwlaat
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Pantep Angchaisuksiri
- Division of Hematology, Department of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | - Kathryn Dane
- Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, MD
| | - Maria T. DeSancho
- Division of Hematology-Oncology, Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY
| | | | - Daniel O. Griffin
- Division of Infectious Diseases, College of Physicians & Surgeons of Columbia University, New York, NY
- Research and Development at United Health Group, Minnetonka, MN
- Prohealth NY, Lake Success, NY
| | - Susan R. Kahn
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Frederikus A. Klok
- Department of Medicine – Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Alfred Ian Lee
- Section of Hematology, Yale School of Medicine, New Haven, CT
| | - Ignacio Neumann
- Department of Internal Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ashok Pai
- Division of Hematology & Oncology, Kaiser Permanente, Oakland/Richmond, CA
| | - Marc Righini
- Division of Angiology and Hemostasis, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | | | - Deborah M. Siegal
- Department of Medicine and the Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | | | - Deirdra R. Terrell
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | | | - Elie A. Akl
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | | | | | - Angela M. Barbara
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Antonio Bognanni
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Mary Boulos
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Romina Brignardello-Petersen
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Rana Charide
- Clinical Research Institute, American University of Beirut, Beirut, Lebanon
| | - Luis E. Colunga-Lozano
- Department of Clinical Medicine, Health Science Center, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Karin Dearness
- Library Services, St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
| | - Andrea J. Darzi
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Heba Hussein
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Cairo, Egypt
| | - Samer G. Karam
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Razan Mansour
- Office of Scientific Affairs and Research, King Hussein Cancer Center, Amman, Jordan
| | - Gian Paolo Morgano
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Rami Z. Morsi
- Department of Neurology, University of Chicago, Chicago, IL
| | - Giovanna Muti-Schünemann
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Menatalla K. Nadim
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Binu A. Philip
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Yuan Qiu
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Yetiani Roldan Benitez
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Adrienne Stevens
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Karla Solo
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Wojtek Wiercioch
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Reem A. Mustafa
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
- Department of Internal Medicine, Division of Nephrology, University of Kansas Medical Center, Kansas City, KS
| | - Holger J. Schünemann
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
- Institut für Evidence in Medicine, Medical Center & Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
115
|
Cuker A, Tseng EK, Schünemann HJ, Angchaisuksiri P, Blair C, Dane K, DeSancho MT, Diuguid D, Griffin DO, Kahn SR, Klok FA, Lee AI, Neumann I, Pai A, Righini M, Sanfilippo KM, Siegal DM, Skara M, Terrell DR, Touri K, Akl EA, Al Jabiri R, Al Jabiri Y, Boulos M, Brignardello-Petersen R, Charide R, Colunga-Lozano LE, Dearness K, Darzi AJ, Karam SG, Morgano GP, Morsi RZ, Philip BA, Benitez YR, Stevens A, Solo K, Wiercioch W, Mustafa RA, Nieuwlaat R. American Society of Hematology living guidelines on the use of anticoagulation for thromboprophylaxis for patients with COVID-19: March 2022 update on the use of anticoagulation in critically ill patients. Blood Adv 2022; 6:4975-4982. [PMID: 35748885 PMCID: PMC9236618 DOI: 10.1182/bloodadvances.2022007940] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/13/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND COVID-19-related critical illness is associated with an increased risk of venous thromboembolism (VTE). OBJECTIVE These evidence-based guidelines of the American Society of Hematology (ASH) are intended to support patients, clinicians, and other health care professionals in decisions about the use of anticoagulation for patients with COVID-19. METHODS ASH formed a multidisciplinary guideline panel, including 3 patient representatives, and applied strategies to minimize potential bias from conflicts of interest. The McMaster University Grading of Recommendations Assessment, Development and Evaluation (GRADE) Centre supported the guideline development process, including performing systematic evidence reviews (up to January 2022). The panel prioritized clinical questions and outcomes according to their importance for clinicians and patients. The panel used the GRADE approach to assess evidence and make recommendations, which were subject to public comment. This is an update to guidelines published in February 2021 and May 2021 as part of the living phase of these guidelines. RESULTS The panel made 1 additional recommendation: a conditional recommendation for the use of prophylactic-intensity over therapeutic-intensity anticoagulation for patients with COVID-19-related critical illness who do not have suspected or confirmed VTE. The panel emphasized the need for an individualized assessment of thrombotic and bleeding risk. CONCLUSIONS This conditional recommendation was based on very low certainty in the evidence, underscoring the need for additional, high-quality, randomized controlled trials comparing different intensities of anticoagulation for patients with COVID-19-related critical illness.
Collapse
Affiliation(s)
- Adam Cuker
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eric K. Tseng
- Division of Hematology/Oncology; St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| | - Holger J. Schünemann
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
- Institut für Evidence in Medicine, Medical Center & Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Pantep Angchaisuksiri
- Division of Hematology, Department of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | - Kathryn Dane
- Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, MD
| | - Maria T. DeSancho
- Division of Hematology-Oncology, Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY
| | | | - Daniel O. Griffin
- Division of Infectious Diseases, College of Physicians & Surgeons of Columbia University, New York, NY
- Research and Development at United Health Group, Minnetonka, MN
- Prohealth NY, Lake Success, NY
| | - Susan R. Kahn
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Frederikus A. Klok
- Department of Medicine–Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Alfred Ian Lee
- Section of Hematology, Yale School of Medicine, New Haven, CT
| | - Ignacio Neumann
- Escuela de Medicina, Facultad de Medicina y Ciencia, Universidad San Sebastian, Santiago, Chile
| | - Ashok Pai
- Division of Hematology & Oncology, Kaiser Permanente, Oakland/Richmond, CA
| | - Marc Righini
- Division of Angiology and Hemostasis, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | | | - Deborah M. Siegal
- Department of Medicine and the Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | | | - Deirdra R. Terrell
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | | | - Elie A. Akl
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | | | | | - Mary Boulos
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Romina Brignardello-Petersen
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Rana Charide
- Clinical Research Institute, American University of Beirut, Beirut, Lebanon
| | - Luis E. Colunga-Lozano
- Department of Clinical Medicine, Health Science Center, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Karin Dearness
- Library Services, St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
| | - Andrea J. Darzi
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Samer G. Karam
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Gian Paolo Morgano
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Rami Z. Morsi
- Department of Neurology, University of Chicago, Chicago, IL; and
| | - Binu A. Philip
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Yetiani Roldan Benitez
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Adrienne Stevens
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Karla Solo
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Wojtek Wiercioch
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| | - Reem A. Mustafa
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
- Department of Internal Medicine, Division of Nephrology, University of Kansas Medical Center, Kansas City, KS
| | - Robby Nieuwlaat
- Department of Health Research Methods, Evidence, and Impact, Michael G. DeGroote Cochrane Canada and GRADE Centres, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
116
|
Dix C, Zeller J, Stevens H, Eisenhardt SU, Shing KSCT, Nero TL, Morton CJ, Parker MW, Peter K, McFadyen JD. C-reactive protein, immunothrombosis and venous thromboembolism. Front Immunol 2022; 13:1002652. [PMID: 36177015 PMCID: PMC9513482 DOI: 10.3389/fimmu.2022.1002652] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/22/2022] [Indexed: 11/24/2022] Open
Abstract
C-reactive protein (CRP) is a member of the highly conserved pentraxin superfamily of proteins and is often used in clinical practice as a marker of infection and inflammation. There is now increasing evidence that CRP is not only a marker of inflammation, but also that destabilized isoforms of CRP possess pro-inflammatory and pro-thrombotic properties. CRP circulates as a functionally inert pentameric form (pCRP), which relaxes its conformation to pCRP* after binding to phosphocholine-enriched membranes and then dissociates to monomeric CRP (mCRP). with the latter two being destabilized isoforms possessing highly pro-inflammatory features. pCRP* and mCRP have significant biological effects in regulating many of the aspects central to pathogenesis of atherothrombosis and venous thromboembolism (VTE), by directly activating platelets and triggering the classical complement pathway. Importantly, it is now well appreciated that VTE is a consequence of thromboinflammation. Accordingly, acute VTE is known to be associated with classical inflammatory responses and elevations of CRP, and indeed VTE risk is elevated in conditions associated with inflammation, such as inflammatory bowel disease, COVID-19 and sepsis. Although the clinical data regarding the utility of CRP as a biomarker in predicting VTE remains modest, and in some cases conflicting, the clinical utility of CRP appears to be improved in subsets of the population such as in predicting VTE recurrence, in cancer-associated thrombosis and in those with COVID-19. Therefore, given the known biological function of CRP in amplifying inflammation and tissue damage, this raises the prospect that CRP may play a role in promoting VTE formation in the context of concurrent inflammation. However, further investigation is required to unravel whether CRP plays a direct role in the pathogenesis of VTE, the utility of which will be in developing novel prophylactic or therapeutic strategies to target thromboinflammation.
Collapse
Affiliation(s)
- Caroline Dix
- Department of Haematology, Alfred Hospital, Melbourne, VIC, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Johannes Zeller
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Plastic and Hand Surgery, University of Freiburg Medical Centre, Medical Faculty of the University of Freiburg, Freiburg, Germany
| | - Hannah Stevens
- Department of Haematology, Alfred Hospital, Melbourne, VIC, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Steffen U. Eisenhardt
- Department of Plastic and Hand Surgery, University of Freiburg Medical Centre, Medical Faculty of the University of Freiburg, Freiburg, Germany
| | - Karen S. Cheung Tung Shing
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, VIC, Australia
| | - Tracy L. Nero
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, VIC, Australia
| | - Craig J. Morton
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, VIC, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Biomedical Manufacturing Program, Clayton, VIC, Australia
| | - Michael W. Parker
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, VIC, Australia
- Structural Biology Unit, St. Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, VIC, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, VIC, Australia
| | - James D. McFadyen
- Department of Haematology, Alfred Hospital, Melbourne, VIC, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, VIC, Australia
- *Correspondence: James D. McFadyen,
| |
Collapse
|
117
|
Venous Thromboembolic Disease in COVID-19, Pathophysiology, Therapy and Prophylaxis. Int J Mol Sci 2022; 23:ijms231810372. [PMID: 36142282 PMCID: PMC9499629 DOI: 10.3390/ijms231810372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 01/08/2023] Open
Abstract
For over two years, the world has been facing the epidemiological and health challenge of the coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Growing problems are also complications after the development of COVID-19 in the form of post and long- COVID syndromes, posing a challenge for the medical community, both for clinicians and the scientific world. SARS-CoV-2 infection is associated with an increased risk of cardiovascular complications, especially thromboembolic complications, which are associated with both thrombosis of small and very small vessels due to immunothrombosis, and the development of venous thromboembolism. Low molecular wight heparin (LMHW) are the basic agents used in the prevention and treatment of thromboembolic complications in COVID-19. There is still a great deal of controversy regarding both the prevention and treatment of thromboembolic complications, including the prophylaxis dose or the optimal duration of anticoagulant treatment in patients with an episode of venous thromboembolism.
Collapse
|
118
|
Abraham S, Manohar SA, Patel R, Saji AM, Dani SS, Ganatra S. Strategies for Cardio-Oncology Care During the COVID-19 Pandemic. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2022; 24:137-153. [PMID: 36090762 PMCID: PMC9446588 DOI: 10.1007/s11936-022-00965-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2022] [Indexed: 12/15/2022]
Abstract
Purpose of review The COVID-19 pandemic has disrupted healthcare and has disproportionately affected the marginalized populations. Patients with cancer and cardiovascular disease (cardio-oncology population) are uniquely affected. In this review, we explore the current data on COVID-19 vulnerability and outcomes in these patients and discuss strategies for cardio-oncology care with a focus on healthcare innovation, health equity, and inclusion. Recent findings The growing evidence suggest increased morbidity and mortality from COVID-19 in patients with comorbid cancer and cardiovascular disease. Additionally, de novo cardiovascular complications such as myocarditis, myocardial infarction, arrhythmia, heart failure, and thromboembolic events have increasingly emerged, possibly due to an accentuated host immune response and cytokine release syndrome. Summary Patient-centric policies are helpful for cardio-oncology surveillance like remote monitoring, increased use of biomarker-based surveillance, imaging modalities like CT scan, and point-of-care ultrasound to minimize the exposure for high-risk patients. Abundant prior experience in cancer therapy scaffolded the repurposed use of corticosteroids, IL-6 inhibitors, and Janus kinase inhibitors in the treatment of COVID-19 infection. COVID-19 vaccine timing and dose frequency present a challenge due to overlapping toxicities and immune cell depletion in patients receiving cancer therapies. The SARS-CoV-2 pandemic laid bare social and ethnic disparities in healthcare but also steered in innovation to combat problems of patient outreach, particularly with virtual care. In the recovery phase, the backlog in cardio-oncology care, interplay of cancer therapy-related side effects, and long COVID-19 syndrome are crucial issues to address.
Collapse
Affiliation(s)
- Sonu Abraham
- Department of Cardiovascular Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805 USA
| | | | - Rushin Patel
- Department of Cardiovascular Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805 USA
| | - Anu Mariam Saji
- Department of Internal Medicine, Saint Vincent Hospital, Worcester, MA USA
| | - Sourbha S. Dani
- Department of Cardiovascular Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805 USA
| | - Sarju Ganatra
- Department of Cardiovascular Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805 USA
| |
Collapse
|
119
|
Corica B, Tartaglia F, D'Amico T, Romiti GF, Cangemi R. Sex and gender differences in community-acquired pneumonia. Intern Emerg Med 2022; 17:1575-1588. [PMID: 35852675 PMCID: PMC9294783 DOI: 10.1007/s11739-022-02999-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022]
Abstract
Awareness of the influence of sex ands gender on the natural history of several diseases is increasing. Community-acquired pneumonia (CAP) is the most common acute respiratory disease, and it is associated with both morbidity and mortality across all age groups. Although a role for sex- and gender-based differences in the development and associated complications of CAP has been postulated, there is currently high uncertainty on the actual contribution of these factors in the epidemiology and clinical course of CAP. More evidence has been produced on the topic during the last decades, and sex- and gender-based differences have also been extensively studied in COVID-19 patients since the beginning of the SARS-CoV-2 pandemic. This review aims to provide an extensive outlook of the role of sex and gender in the epidemiology, pathogenesis, treatment, and outcomes of patients with CAP, and on the future research scenarios, with also a specific focus on COVID-19.
Collapse
Affiliation(s)
- Bernadette Corica
- Department of Translational and Precision Medicine, Sapienza-University of Rome, Viale del Policlinico 155, 00162, Rome, Italy
| | - Francesco Tartaglia
- Department of Translational and Precision Medicine, Sapienza-University of Rome, Viale del Policlinico 155, 00162, Rome, Italy
| | - Tania D'Amico
- Department of Translational and Precision Medicine, Sapienza-University of Rome, Viale del Policlinico 155, 00162, Rome, Italy
| | - Giulio Francesco Romiti
- Department of Translational and Precision Medicine, Sapienza-University of Rome, Viale del Policlinico 155, 00162, Rome, Italy
| | - Roberto Cangemi
- Department of Translational and Precision Medicine, Sapienza-University of Rome, Viale del Policlinico 155, 00162, Rome, Italy.
| |
Collapse
|
120
|
Butt A, Erkan D, Lee AI. COVID-19 and antiphospholipid antibodies. Best Pract Res Clin Haematol 2022; 35:101402. [PMID: 36494152 PMCID: PMC9568270 DOI: 10.1016/j.beha.2022.101402] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 12/14/2022]
Abstract
Antiphospholipid syndrome and the coagulopathy of COVID-19 share many pathophysiologic features, including endotheliopathy, hypercoagulability, and activation of platelets, complement pathways, and neutrophil extracellular traps, all acting in concert via a model of immunothrombosis. Antiphospholipid antibody production in COVID-19 is common, with 50% of COVID-19 patients being positive for lupus anticoagulant in some studies, and with non-Sapporo criteria antiphospholipid antibodies being prevalent as well. The biological significance of antiphospholipid antibodies in COVID-19 is uncertain, as such antibodies are usually transient, and studies examining clinical outcomes in COVID-19 patients with and without antiphospholipid antibodies have yielded conflicting results. In this review, we explore the biology of antiphospholipid antibodies in COVID-19 and other infections and discuss mechanisms of thrombogenesis in antiphospholipid syndrome and parallels with COVID-19 coagulopathy. In addition, we review the existing literature on safety of COVID-19 vaccination in patients with antiphospholipid antibodies and antiphospholipid syndrome.
Collapse
Affiliation(s)
- Ayesha Butt
- Section of Hematology, Department of Medicine, Yale School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA.
| | - Doruk Erkan
- Barbara Volcker Center for Women and Rheumatic Diseases, Hospital for Special Surgery and Weill Cornell Medicine, 535 E. 70th St., 6th floor, New York, NY, 10021, USA.
| | - Alfred Ian Lee
- Section of Hematology, Department of Medicine, Yale School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA.
| |
Collapse
|
121
|
Panichpisal K, Ruff I, Singh M, Hamidi M, Salinas PD, Swanson K, Medlin S, Dandapat S, Tepp P, Kuchinsky G, Pesch A, Wolfe T. Cerebral Venous Sinus Thrombosis Associated With Coronavirus Disease 2019: Case Report and Review of the Literature. Neurologist 2022; 27:253-262. [PMID: 34855659 PMCID: PMC9439631 DOI: 10.1097/nrl.0000000000000390] [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] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Coronavirus disease 2019 (COVID-19) is associated with significant risk of acute thrombosis. We present a case report of a patient with cerebral venous sinus thrombosis (CVST) associated with COVID-19 and performed a literature review of CVST associated with COVID-19 cases. CASE REPORT A 38-year-old woman was admitted with severe headache and acute altered mental status a week after confirmed diagnosis of COVID-19. Magnetic resonance imaging brain showed diffuse venous sinus thrombosis involving the superficial and deep veins, and diffuse edema of bilateral thalami, basal ganglia and hippocampi because of venous infarction. Her neurological exam improved with anticoagulation (AC) and was subsequently discharged home. We identified 43 patients presenting with CVST associated with COVID-19 infection. 56% were male with mean age of 51.8±18.2 years old. The mean time of CVST diagnosis was 15.6±23.7 days after onset of COVID-19 symptoms. Most patients (87%) had thrombosis of multiple dural sinuses and parenchymal changes (79%). Almost 40% had deep cerebral venous system thrombosis. Laboratory findings revealed elevated mean D-dimer level (7.14/mL±12.23 mg/L) and mean fibrinogen level (4.71±1.93 g/L). Less than half of patients had prior thrombotic risk factors. Seventeen patients (52%) had good outcomes (mRS <=2). The mortality rate was 39% (13 patients). CONCLUSION CVST should be in the differential diagnosis when patients present with acute neurological symptoms in this COVID pandemic. The mortality rate of CVST associated with COVID-19 can be very high, therefore, early diagnosis and prompt treatment are crucial to the outcomes of these patients.
Collapse
Affiliation(s)
| | - Ilana Ruff
- Aurora Neurosciences Innovative Institute
| | - Maharaj Singh
- School of Dentistry, Marquette University
- Aurora Research Institute, Milwaukee, WI
| | | | - Pedro D. Salinas
- Aurora Critical Care Services, Aurora Sinai/Aurora St. Luke’s Medical Centers, University of Wisconsin School of Medicine and Public Health
| | | | | | | | | | | | - Amy Pesch
- Aurora Neurosciences Innovative Institute
| | | |
Collapse
|
122
|
Increased Susceptibility for Thromboembolic Events versus High Bleeding Risk Associated with COVID-19. Microorganisms 2022; 10:microorganisms10091738. [PMID: 36144340 PMCID: PMC9505654 DOI: 10.3390/microorganisms10091738] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
The infection with the SARS-CoV-2 virus is associated with numerous systemic involvements. Besides the severe respiratory injuries and cardiovascular complications, it became obvious early on that this disease carries an increased risk of thromboembolic events, but a higher propensity for bleedings as well. We researched the medical literature over significant PubMed published articles debating on the prevalence, category of patients, the moment of occurrence, and evolution of venous thromboembolism (VTE), but also of venous and arterial “in situ” thrombosis (AT), and hemorrhagic events as well. Most researchers agree on an increased prevalence of thromboembolic events, ranging between 25 and 31% for VTE, depending on the analyzed population. For AT and hemorrhagic complications lower rates were reported, namely, about 2–3%, respectively, between 4.8 and 8%, occurring mostly in older patients, suffering from moderate/severe forms of COVID-19, with associated comorbidities. It is important to mention that patients suffering from hemorrhages frequently received thromboprophylaxis with anticoagulant drugs. As a consequence of thromboembolic and hemorrhagic complications which are both important negative prognostic factors, the evolution of patients infected with the SARS-CoV-2 virus is aggravated, determining an augmented morbidity and mortality of this population.
Collapse
|
123
|
Danis F, Rabani AE, Subhani F, Yasmin S, Koul SS. COVID‐19: Vaccine‐induced immune thrombotic thrombocytopenia (VITT). Eur J Haematol 2022; 109:619-632. [PMID: 36030503 PMCID: PMC9538855 DOI: 10.1111/ejh.13855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
Affiliation(s)
| | | | | | | | - Salman Shafi Koul
- Pakistan Institute of Medical Sciences, Ibn‐e‐Sina Road, G‐8/3 G 8/3 G‐8 Islamabad Pakistan
| |
Collapse
|
124
|
Voigtlaender M, Edler C, Gerling M, Schädler J, Ondruschka B, Schröder AS, Sperhake J, Ehrhardt S, Wang L, Haddad M, Kiencke V, Renné T, Roedl K, Kluge S, Wichmann D, Langer F. Thromboembolic events in deceased patients with proven SARS-CoV-2 infection: Frequency, characteristics and risk factors. Thromb Res 2022; 218:171-176. [PMID: 36057167 PMCID: PMC9420077 DOI: 10.1016/j.thromres.2022.08.021] [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: 03/26/2022] [Revised: 08/11/2022] [Accepted: 08/19/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Infection with the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) results in respiratory syndromes but also in vascular complications such as thromboembolism (TE). In this regard, immunothrombosis, resulting from inflammation in SARS-CoV-2 infected tissues, has been described. Data on TE in COVID-19 are mainly based on clinical observational and/or incomplete autopsy studies. The true burden of TE and the relevance of genetic predisposition, however, have not been resolved. OBJECTIVES Here, we report on a consecutive cohort of 100 fully autopsied patients deceased by SARS-CoV-2 infections during the first wave of the pandemic (March to April 2020). We investigated the localization of TE, potential clinical risk factors, and the prothrombotic gene mutations, factor V Leiden and prothrombin G20210A, in postmortem blood or tissue samples. RESULTS TE was found in 43/100 autopsies. 93 % of TE events were venous occlusions, with 23 patients having pulmonary thromboembolism (PT) with or without lower-extremity deep vein thrombosis. Of these, 70 % showed PT restricted to (sub)segmental arteries, consistent with in situ immunothrombosis. Patients with TE had a significantly higher BMI and died more frequently at an intensive care unit. Hereditary thrombophilia factors were not associated with TE. CONCLUSIONS Our autopsy results show that a significant proportion of SARS-CoV-2 infected patients suffer from TE, affecting predominantly the venous system. Orthotopic peripheral PT was the most frequent finding. Hereditary thrombophilia appears not to be a determinant for TE in COVID-19. However, obesity and the need for intensive care increase the risk of TE in these patients.
Collapse
Affiliation(s)
- Minna Voigtlaender
- II. Medical Department - Oncology, Hematology, Bone Marrow Transplantation and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carolin Edler
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Moritz Gerling
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Schädler
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ann Sophie Schröder
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Sperhake
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Ehrhardt
- Bloomberg School of Public Health, Department of Epidemiology, Johns Hopkins University Baltimore, Baltimore, USA
| | - Lin Wang
- Bloomberg School of Public Health, Department of Epidemiology, Johns Hopkins University Baltimore, Baltimore, USA
| | - Munif Haddad
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Verena Kiencke
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland; Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Kevin Roedl
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dominic Wichmann
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Langer
- II. Medical Department - Oncology, Hematology, Bone Marrow Transplantation and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
125
|
Russo V, Caputo A, Imbalzano E, Di Micco P, Frontera A, Uccello A, Orlando L, Galimberti P, Golino P, D'Andrea A. The pharmacology of anticoagulant drug treatment options in COVID-19 patients: reviewing real-world evidence in clinical practice. Expert Rev Clin Pharmacol 2022; 15:1095-1105. [PMID: 36017645 DOI: 10.1080/17512433.2022.2117154] [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: 11/04/2022]
Abstract
INTRODUCTION The optimal anticoagulation strategy for venous thromboembolism (VTE) prevention among COVID-19 patients, hospitalized or in the community setting, is still challenging and largely based on real-world evidence. AREAS COVERED We analyzed real-world data regarding the safety and effectiveness of anticoagulant treatment, both parenteral and oral, for VTE prevention or atrial fibrillation (AF)/VTE treatment among COVID-19 patients. EXPERT OPINION The efficacy of low-molecular-weight heparin (LMWH) doses for VTE prevention correlates with COVID-19 disease status. LMWH prophylactic dose may be useful in COVID-19 patients at the early stage of the disease. LMWH intermediate or therapeutic dose is recommended in COVID-19 patients with an advanced stage of the disease. COVID-19 patients on VKAs therapy for atrial fibrillation (AF) and VTE should switch to NOACs in the community setting or LMWH in the hospital setting. No definitive data on de-novo starting of NOACs or VKAs therapy for VTE prevention in COVID-19 outpatients are available. In patients at high risk discharged after hospitalization due to COVID-19, thromboprophylaxis with NOACs may be considered.
Collapse
Affiliation(s)
- Vincenzo Russo
- Department of Medical Translational Sciences, University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy
| | - Adriano Caputo
- Department of Medical Translational Sciences, University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy
| | - Egidio Imbalzano
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Pierpaolo Di Micco
- Department of Internal Medicine and Cardiology, Fatebenefratelli Hospital, Naples, Italy
| | - Antonio Frontera
- Arrhythmology Department, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Ambra Uccello
- Department of Internal Medicine and Cardiology, Fatebenefratelli Hospital, Naples, Italy
| | - Luana Orlando
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Paola Galimberti
- Arrhythmology Department, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Paolo Golino
- Department of Medical Translational Sciences, University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy
| | - Antonello D'Andrea
- Department of Cardiology, Umberto I Hospital, 84014 Nocera Inferiore, Italy
| |
Collapse
|
126
|
Antic D, Milic N, Chatzikonstantinou T, Scarfò L, Otasevic V, Rajovic N, Allsup D, Alonso Cabrero A, Andres M, Baile Gonzales M, Capasso A, Collado R, Cordoba R, Cuéllar-García C, Correa JG, De Paoli L, De Paolis MR, Del Poeta G, Dimou M, Doubek M, Efstathopoulou M, El-Ashwah S, Enrico A, Espinet B, Farina L, Ferrari A, Foglietta M, Lopez-Garcia A, García-Marco JA, García-Serra R, Gentile M, Gimeno E, da Silva MG, Gutwein O, Hakobyan YK, Herishanu Y, Hernández-Rivas JÁ, Herold T, Itchaki G, Jaksic O, Janssens A, Kalashnikova OB, Kalicińska E, Kater AP, Kersting S, Koren-Michowitz M, Labrador J, Lad D, Laurenti L, Fresa A, Levin MD, Mayor Bastida C, Malerba L, Marasca R, Marchetti M, Marquet J, Mihaljevic B, Milosevic I, Mirás F, Morawska M, Motta M, Munir T, Murru R, Nunes R, Olivieri J, Pavlovsky MA, Piskunova I, Popov VM, Quaglia FM, Quaresmini G, Reda G, Rigolin GM, Shrestha A, Šimkovič M, Smirnova S, Špaček M, Sportoletti P, Stanca O, Stavroyianni N, Te Raa D, Tomic K, Tonino S, Trentin L, Van Der Spek E, van Gelder M, Varettoni M, Visentin A, Vitale C, Vukovic V, Wasik-Szczepanek E, Wróbel T, Segundo LYS, Yassin M, Coscia M, Rambaldi A, Montserrat E, Foà R, Cuneo A, Carrier M, Ghia P, Stamatopoulos K. Thrombotic and bleeding complications in patients with chronic lymphocytic leukemia and severe COVID-19: a study of ERIC, the European Research Initiative on CLL. J Hematol Oncol 2022; 15:116. [PMID: 36028857 PMCID: PMC9415249 DOI: 10.1186/s13045-022-01333-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/08/2022] [Indexed: 12/03/2022] Open
Abstract
Background Patients with chronic lymphocytic leukemia (CLL) may be more susceptible to COVID-19 related poor outcomes, including thrombosis and death, due to the advanced age, the presence of comorbidities, and the disease and treatment-related immune deficiency. The aim of this study was to assess the risk of thrombosis and bleeding in patients with CLL affected by severe COVID-19. Methods This is a retrospective multicenter study conducted by ERIC, the European Research Initiative on CLL, including patients from 79 centers across 22 countries. Data collection was conducted between April and May 2021. The COVID-19 diagnosis was confirmed by the real-time polymerase chain reaction (RT-PCR) assay for SARS-CoV-2 on nasal or pharyngeal swabs. Severe cases of COVID-19 were defined by hospitalization and the need of oxygen or admission into ICU. Development and type of thrombotic events, presence and severity of bleeding complications were reported during treatment for COVID-19. Bleeding events were classified using ISTH definition. STROBE recommendations were used in order to enhance reporting. Results A total of 793 patients from 79 centers were included in the study with 593 being hospitalized (74.8%). Among these, 511 were defined as having severe COVID: 162 were admitted to the ICU while 349 received oxygen supplementation outside the ICU. Most patients (90.5%) were receiving thromboprophylaxis. During COVID-19 treatment, 11.1% developed a thromboembolic event, while 5.0% experienced bleeding. Thrombosis developed in 21.6% of patients who were not receiving thromboprophylaxis, in contrast to 10.6% of patients who were on thromboprophylaxis. Bleeding episodes were more frequent in patients receiving intermediate/therapeutic versus prophylactic doses of low-molecular-weight heparin (LWMH) (8.1% vs. 3.8%, respectively) and in elderly. In multivariate analysis, peak D-dimer level and C-reactive protein to albumin ratio were poor prognostic factors for thrombosis occurrence (OR = 1.022, 95%CI 1.007‒1.038 and OR = 1.025, 95%CI 1.001‒1.051, respectively), while thromboprophylaxis use was protective (OR = 0.199, 95%CI 0.061‒0.645). Age and LMWH intermediate/therapeutic dose administration were prognostic factors in multivariate model for bleeding (OR = 1.062, 95%CI 1.017–1.109 and OR = 2.438, 95%CI 1.023–5.813, respectively). Conclusions Patients with CLL affected by severe COVID-19 are at a high risk of thrombosis if thromboprophylaxis is not used, but also at increased risk of bleeding under the LMWH intermediate/therapeutic dose administration.
Collapse
Affiliation(s)
- Darko Antic
- Lymphoma Center, Clinic for Hematology, University Clinical Center of Serbia, Belgrade, Serbia. .,Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
| | - Natasa Milic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Thomas Chatzikonstantinou
- Hematology Department and HCT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece.,Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Lydia Scarfò
- Università Vita-Salute San Raffaele and IRCC Ospedale San Raffaele, Milan, Italy
| | - Vladimir Otasevic
- Lymphoma Center, Clinic for Hematology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Nina Rajovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - David Allsup
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, Hull, UK
| | | | - Martin Andres
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | | | - Rosa Collado
- Department of Hematology, Hospital General Universitario, Valencia, Spain.,Fundación de Investigación del Hospital General Universitario, Valencia, Spain
| | - Raul Cordoba
- Department of Hematology, Health Research Institute IIS-FJD, Fundacion Jimenez Diaz University Hospital, Madrid, Spain
| | | | | | - Lorenzo De Paoli
- Division of Internal Medicine, Hematology Unit, ASL Vercelli, Vercelli, Italy
| | | | - Giovanni Del Poeta
- Department of Biomedicine and Prevention Hematology, University Tor Vergata, Rome, Italy
| | - Maria Dimou
- 1st Internal Medicine Department, Propaedeutic, Hematology Clinical Trial Unit, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael Doubek
- Department of Internal Medicine - Hematology and Oncology, University Hospital, Brno, Czechia.,Department of Medical Genetics and Genomics, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Maria Efstathopoulou
- Department of Haematology Athens Medical Center-Psychikon Branch, Athens, Greece
| | - Shaimaa El-Ashwah
- Clinical Hematology Unit, Oncology Center, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | | | - Blanca Espinet
- Department of Hematology, Hospital del Mar, Barcelona, Spain
| | - Lucia Farina
- Hematology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Angela Ferrari
- Hematology Unit, Azienda Unità Sanitaria Locale - IRCCS, Reggio Emilia, Italy
| | | | - Alberto Lopez-Garcia
- Department of Hematology, Health Research Institute IIS-FJD, Fundacion Jimenez Diaz University Hospital, Madrid, Spain
| | - José A García-Marco
- Hematology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Rocío García-Serra
- Department of Hematology, Hospital General Universitario, Valencia, Spain.,Fundación de Investigación del Hospital General Universitario, Valencia, Spain
| | | | - Eva Gimeno
- Department of Hematology, Hospital del Mar, Barcelona, Spain
| | - Maria Gomes da Silva
- Hematology Department, Portuguese Institute of Oncology Lisbon, Lisbon, Portugal
| | - Odit Gutwein
- Department of Hematology, Shamir Medical Center, Zerifin, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Yair Herishanu
- Department of Hematology, Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Tobias Herold
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Gilad Itchaki
- Division of Hematology, Rabin Medical Center, Petah Tikva, and the Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ozren Jaksic
- Department of Hematology, University Hospital Dubrava, Zagreb, Croatia
| | - Ann Janssens
- Department of Hematology, Universitaire Ziekenhuizen Leuven, Leuven, Belgium
| | - Olga B Kalashnikova
- Federal State Budgetary Educational Institution of Higher Education Academician I.P. Pavlov, First St. Petersburg State Medical University of the Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Elżbieta Kalicińska
- Department and Clinic of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Pasteura Street 4, 50-367, Wrocław, Poland
| | - Arnon P Kater
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Sabina Kersting
- Department of Hematology, Haga Teaching Hospital, The Hague, The Netherlands
| | - Maya Koren-Michowitz
- Department of Hematology, Shamir Medical Center, Zerifin, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jorge Labrador
- Hematology Department, Unit Research, Complejo Asistencial Universitario de Burgos, Burgos, Spain
| | - Deepesh Lad
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Luca Laurenti
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy.,Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alberto Fresa
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy.,Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Mark-David Levin
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Carlota Mayor Bastida
- Haematology Department, Hospital Universitario de La Princesa, Madrid, Spain.,Spanish Society of Haematology and Hemotherapy (SEHH: Sociedad Española de Hematología y Hemoterapia), Madrid, Spain
| | - Lara Malerba
- Hematology and Stem Cell Transplant Center Marche Nord Hospital, Pesaro, Italy
| | - Roberto Marasca
- Section of Hematology, Department of Medical Sciences, University of Modena and Reggio E., Modena, Italy
| | - Monia Marchetti
- Hematology Unit & TMO Center, AO SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Juan Marquet
- Hematology Department, Ramón y Cajal University Hospital, Madrid, Spain
| | - Biljana Mihaljevic
- Lymphoma Center, Clinic for Hematology, University Clinical Center of Serbia, Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ivana Milosevic
- Clinical Centre of Vojvodina, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Fatima Mirás
- Hematology Department, Hospital Universitario 12de Octubre, Madrid, Spain
| | - Marta Morawska
- Experimental Hematooncology Department, Medical University of Lublin, Lublin, Poland.,Hematology Department, St. John's Cancer Center, Lublin, Poland
| | - Marina Motta
- S.C. Ematologia ASST Spedali Civili Brescia, Brescia, Italy
| | - Talha Munir
- Consultant Haematologist, St James's Hospital, Leeds, LS9 7TF, UK
| | - Roberta Murru
- Hematology and Stem Cell Transplantation Unit, Ospedale Oncologico A. Businco, ARNAS "G. Brotzu", Cagliari, Italy
| | - Raquel Nunes
- Hematology Department, Portuguese Institute of Oncology Lisbon, Lisbon, Portugal
| | | | | | - Inga Piskunova
- Consultative Hematology Department with a Day Hospital for Intensive High-Dose Chemotherapy, National Research Center for Hematology, Moscow, Russia
| | - Viola Maria Popov
- Hematology Department, Colentina Clinical Hospital, Bucharest, Romania
| | | | | | - Gianluigi Reda
- Hematology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico of Milan, Milan, Italy
| | | | - Amit Shrestha
- Hematology Unit, Nepal Cancer Hospital and Research Center, Lalitpur, Nepal
| | - Martin Šimkovič
- 4th Department of Internal Medicine - Haematology, Faculty of Medicine in Hradec Králové, University Hospital and Charles University in Prague, Hradec Kralove, Czech Republic
| | - Svetlana Smirnova
- Consultative Hematology Department with a Day Hospital for Intensive High-Dose Chemotherapy, National Research Center for Hematology, Moscow, Russia
| | - Martin Špaček
- 1st Department of Medicine - Hematology, First Faculty of Medicine, Charles University and General Hospital in Prague, Prague, Czech Republic
| | - Paolo Sportoletti
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncological Research, Ospedale S. Maria della Misericordia, Perugia, Italy
| | - Oana Stanca
- Hematology Department from Coltea Clinical Hospital, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Niki Stavroyianni
- Hematology Department and HCT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| | - Doreen Te Raa
- Department of Hematology, Gelderse Vallei Ede, Ede, The Netherlands
| | - Kristina Tomic
- Lymphoma Center, Clinic for Hematology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Sanne Tonino
- Department of Hematology, Lymmcare, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Livio Trentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padova, Padua, Italy
| | - Ellen Van Der Spek
- Department of Internal Medicine, Rijnstate Hospital, Arnhem, The Netherlands
| | - Michel van Gelder
- Department Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Marzia Varettoni
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Andrea Visentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padova, Padua, Italy
| | - Candida Vitale
- Division of Hematology, A.O.U. Cittàdella Salute e della Scienza di Torino and Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Vojin Vukovic
- Lymphoma Center, Clinic for Hematology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Ewa Wasik-Szczepanek
- Department Hematooncology and Bone Marrow Transplantation, Medical University in Lublin, Lublin, Poland
| | - Tomasz Wróbel
- Department and Clinic of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Pasteura Street 4, 50-367, Wrocław, Poland
| | | | - Mohamed Yassin
- Hematology Section, Department of Medical Oncology, National Center for Cancer Care and Research, Doha, Qatar
| | - Marta Coscia
- Division of Hematology, A.O.U. Cittàdella Salute e della Scienza di Torino and Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | | | | | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | | | - Marc Carrier
- Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, K1H 8L6, Canada
| | - Paolo Ghia
- Università Vita-Salute San Raffaele and IRCC Ospedale San Raffaele, Milan, Italy
| | - Kostas Stamatopoulos
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| |
Collapse
|
127
|
Islam A, Cockcroft C, Elshazly S, Ahmed J, Joyce K, Mahfuz H, Islam T, Rashid H, Laher I. Coagulopathy of Dengue and COVID-19: Clinical Considerations. Trop Med Infect Dis 2022; 7:tropicalmed7090210. [PMID: 36136621 PMCID: PMC9500638 DOI: 10.3390/tropicalmed7090210] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 12/04/2022] Open
Abstract
Thrombocytopenia and platelet dysfunction commonly occur in both dengue and COVID-19 and are related to clinical outcomes. Coagulation and fibrinolytic pathways are activated during an acute dengue infection, and endothelial dysfunction is observed in severe dengue. On the other hand, COVID-19 is characterised by a high prevalence of thrombotic complications, where bleeding is rare and occurs only in advanced stages of critical illness; here thrombin is the central mediator that activates endothelial cells, and elicits a pro-inflammatory reaction followed by platelet aggregation. Serological cross-reactivity may occur between COVID-19 and dengue infection. An important management aspect of COVID-19-induced immunothrombosis associated with thrombocytopenia is anticoagulation with or without aspirin. In contrast, the use of aspirin, nonsteroidal anti-inflammatory drugs and anticoagulants is contraindicated in dengue. Mild to moderate dengue infections are treated with supportive therapy and paracetamol for fever. Severe infection such as dengue haemorrhagic fever and dengue shock syndrome often require escalation to higher levels of support in a critical care facility. The role of therapeutic platelet transfusion is equivocal and should not be routinely used in patients with dengue with thrombocytopaenia and mild bleeding. The use of prophylactic platelet transfusion in dengue fever has strained financial and healthcare systems in endemic areas, together with risks of transfusion-transmitted infections in low- and middle-income countries. There is a clear research gap in the management of dengue with significant bleeding.
Collapse
Affiliation(s)
- Amin Islam
- Department of Haematology, Mid & South Essex University Hospital NHS Foundation Trust, Prittlewell Chase, Westcliff-on-Sea SS0 0RY, UK
- Department of Haematology, Queen Mary University of London, Mile End Road, London E1 3NS, UK
- Correspondence:
| | - Christopher Cockcroft
- Department of Haematology, Mid & South Essex University Hospital NHS Foundation Trust, Prittlewell Chase, Westcliff-on-Sea SS0 0RY, UK
| | - Shereen Elshazly
- Department of Haematology, Mid & South Essex University Hospital NHS Foundation Trust, Prittlewell Chase, Westcliff-on-Sea SS0 0RY, UK
- Adult Haemato-Oncology Unit, Faculty of Medicine, Ainshams University, Cairo 11566, Egypt
| | - Javeed Ahmed
- Department of Microbiology and Virology, Mid & South Essex University Hospital NHS Foundation Trust, Westcliff-on-Sea SS0 0RY, UK
| | - Kevin Joyce
- Department of Haematology, Mid & South Essex University Hospital NHS Foundation Trust, Prittlewell Chase, Westcliff-on-Sea SS0 0RY, UK
| | - Huque Mahfuz
- Department of Haematology and Oncology, Combined Military Hospital, Dhaka 1206, Bangladesh
| | - Tasbirul Islam
- Department of Pulmonology and Critical Care Medicine, Indiana School of Medicine, Lafayette, IN 47907, USA
| | - Harunor Rashid
- National Centre for Immunisation Research and Surveillance, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
- Sydney Institute for Infectious Diseases, The University of Sydney, Westmead, NSW 2145, Australia
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, The University of British Colombia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
128
|
Anticoagulation in hospitalized patients with COVID-19. Blood 2022; 140:809-814. [PMID: 35653590 PMCID: PMC9361053 DOI: 10.1182/blood.2021014527] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/18/2022] [Indexed: 11/20/2022] Open
Abstract
Coronavirus disease-19 (COVID-19) includes a thromboinflammatory syndrome that may manifest with microvascular and macrovascular thrombosis. Patients with COVID-19 have a higher incidence of venous thromboembolism than other hospitalized patients. Three randomized control trials suggesting benefit of therapeutic heparin in hospitalized noncritically ill patients with COVID-19 have led to conditional guideline recommendations for this treatment. By contrast, prophylactic-dose heparin is recommended for critically ill patients. Unprecedented collaboration and rapidly funded research have improved care of hospitalized patients with COVID-19.
Collapse
|
129
|
Alessandro B, Wilma B, Bruno F. Pulmonary embolism in a patient with eltrombopag-treated aplastic anaemia and paroxysmal nocturnal haemoglobinuria clone during COVID-19 pneumonia. Thromb J 2022; 20:46. [PMID: 35999631 PMCID: PMC9398496 DOI: 10.1186/s12959-022-00407-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/26/2022] [Indexed: 12/22/2022] Open
Abstract
Thrombosis in patients with thrombocytopenia has several risk factors, both disease-related and treatment-associated. Recently, COVID-19 infection was recognized as an additional risk factor, further complicating the delicate balance between thrombosis and bleeding in these patients. Here we describe the case of a patient with aplastic anaemia on eltrombopag who developed pulmonary embolism during COVID-19 pneumonia, despite receiving oral anticoagulation with edoxaban. Notably, he was also carrying a large paroxysmal nocturnal haemoglobinuria clone, although without evidence of haemolysis. The presented case recapitulates some of the open questions in thrombotic risk management of cytopenic patients, such as the management of thrombopoietin receptor agonists and the choice of anticoagulation in PNH, while also accounting for the additional thrombotic risk linked to COVID-19.
Collapse
Affiliation(s)
- Bosi Alessandro
- Haematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20100, Milan, Italy.,Department of Oncology and Haemato-oncology, University of Milan, Milan, Italy
| | - Barcellini Wilma
- Haematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20100, Milan, Italy
| | - Fattizzo Bruno
- Haematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20100, Milan, Italy. .,Department of Oncology and Haemato-oncology, University of Milan, Milan, Italy.
| |
Collapse
|
130
|
The prevalence of vascular complications in SARS-CoV-2 infected outpatients. Wien Med Wochenschr 2022; 173:168-172. [PMID: 35939218 PMCID: PMC9358629 DOI: 10.1007/s10354-022-00954-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022]
Abstract
Background The risk of thromboembolic events is increased for coronavirus disease (COVID)-19 inpatients. For severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected outpatients, only few data are available so far. Methods In our prospective single-center study, 461 SARS-CoV-2-infected outpatients were screened for the presence of deep vein thrombosis. Results Two outpatients had suffered a deep vein thrombosis. An association with previously known risk factors, such as preexisting thrombosis in the medical history or cardiovascular risk factors, could not be proven. Conclusion General thromboprophylaxis in SARS-CoV-2-infected outpatients is still not recommended.
Collapse
|
131
|
Falcinelli E, Petito E, Gresele P. The role of platelets, neutrophils and endothelium in COVID-19 infection. Expert Rev Hematol 2022; 15:727-745. [PMID: 35930267 DOI: 10.1080/17474086.2022.2110061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION COVID-19 is associated to an increased risk of thrombosis, as a result of a complex process that involves the activation of vascular and circulating cells, the release of soluble inflammatory and thrombotic mediators and blood clotting activation. AREAS COVERED This article reviews the pathophysiological role of platelets, neutrophils and the endothelium, and of their interactions, in the thrombotic complications of COVID-19 patients, and the current and future therapeutic approaches targeting these cell types. EXPERT OPINION Virus-induced platelet, neutrophil and endothelial cell changes are crucial triggers of the thrombotic complications and of the adverse evolution of COVID-19. Both the direct interaction with the virus and the associated cytokine storm concur to trigger cell activation in a classical thromboinflammatory vicious circle. Although heparin has proven to be an effective prophylactic and therapeutic weapon for the prevention and treatment of COVID-19-associated thrombosis, it acts downstream of the cascade of events triggered by SARS-CoV-2. The identification of specific molecular targets interrupting the thromboinflammatory cascade upstream, and more specifically acting either on the interaction of SARS-CoV-2 with blood and vascular cells or on the specific signalling mechanisms associated with their COVID-19-associated activation, might theoretically offer greater protection with potentially lesser side effects.
Collapse
Affiliation(s)
- E Falcinelli
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - E Petito
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - P Gresele
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
132
|
Hammami R, Jdidi J, Chakroun O, Issaoui F, Ktata N, Maamri H, Baklouti M, Bahloul A, Gargouri R, Nasri A, Msaad S, Kammoun S, Kammoun S, Rejab IB, Charfeddine S, Abid L. Thromboembolic events in COVID-19 ambulatory patients: An observational study about incidence, and thromboprophylaxis outcomes. PLoS One 2022; 17:e0270195. [PMID: 35925930 PMCID: PMC9352084 DOI: 10.1371/journal.pone.0270195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 06/06/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction
There are no clear data about the incidence and the prophylactic strategies of arterial and venous thromboembolic events (TE) in COVID-19 ambulatory patients. Thus, we conducted this study to analyze thromboembolic complications in this setting and to assess thromboprophylaxis management and outcomes in the real life.
Patients and methods
This is an observational study including Covid-19 ambulatory patients. We assessed incidence of venous and arterial TE events as well as thromboprophylaxis outcomes and hemorrhagic complications. We defined high risk thrombo-embolic factor according to the Belgian guidelines which are the only guidelines that described thromboprophylaxis in COVID-19 ambulatory patients.
Results
We included 2089 patients with a mean age of 43±16 years. The incidence of 30 days venous and arterial TE complications in our cohort was 1%. Venous thromboembolic complications occurred in 0.8% and arterial thromboembolic complications occurred in 0.3%.We noted at least one high-risk TE factor in 18.5% of patients but thromboprophylaxis was prescribed in 22.5% of the cases, LMWH in 18.1%, and Rivaroxaban in 3.7%. Hemorrhagic events occurred in eight patients (0.3%): five patients showed minor hemorrhagic events and three patients showed major ones (0.14%).
Conclusions
Our study showed that the incidence of thromboembolic complications is very low in COVID-19 ambulatory patients. Paradoxically, there is an over prescription of thrombo-prophylaxis in this population.
Collapse
Affiliation(s)
- Rania Hammami
- Cardiology Department, Hedi Chaker Hospital, University of Medicine, Sfax University, Sfax, Tunisia
- * E-mail:
| | - Jihen Jdidi
- Epidemiology Department Hedi Chaker Hospital, University of Medicine, Sfax University, Sfax, Tunisia
| | - Olfa Chakroun
- Emergency Department, University Hospital Habib Bourguiba Sfax, University of Medicine, Sfax University, Sfax, Tunisia
| | - Fadhila Issaoui
- Emergency Department, University Hospital Habib Bourguiba Sfax, University of Medicine, Sfax University, Sfax, Tunisia
| | - Nouha Ktata
- Epidemiology Department Hedi Chaker Hospital, University of Medicine, Sfax University, Sfax, Tunisia
| | - Hanen Maamri
- Epidemiology Department Hedi Chaker Hospital, University of Medicine, Sfax University, Sfax, Tunisia
| | - Mouna Baklouti
- Epidemiology Department Hedi Chaker Hospital, University of Medicine, Sfax University, Sfax, Tunisia
| | - Amine Bahloul
- Cardiology Department, Hedi Chaker Hospital, University of Medicine, Sfax University, Sfax, Tunisia
| | - Rania Gargouri
- Cardiology Department, Hedi Chaker Hospital, University of Medicine, Sfax University, Sfax, Tunisia
| | - Abdennour Nasri
- Emergency Department, University Hospital Habib Bourguiba Sfax, University of Medicine, Sfax University, Sfax, Tunisia
| | - Sameh Msaad
- Pneumology department, Hedi Chaker Hospital, University of Medicine, Sfax University, Sfax, Tunisia
| | - Samy Kammoun
- Pneumology department, Hedi Chaker Hospital, University of Medicine, Sfax University, Sfax, Tunisia
| | - Samir Kammoun
- Cardiology Department, Hedi Chaker Hospital, University of Medicine, Sfax University, Sfax, Tunisia
| | | | - Selma Charfeddine
- Cardiology Department, Hedi Chaker Hospital, University of Medicine, Sfax University, Sfax, Tunisia
| | - Leila Abid
- Cardiology Department, Hedi Chaker Hospital, University of Medicine, Sfax University, Sfax, Tunisia
| |
Collapse
|
133
|
Pilia E, Belletti A, Fresilli S, Finco G, Landoni G. Efficacy and safety of heparin full-dose anticoagulation in hospitalized non-critically ill COVID-19 patients: a meta-analysis of multicenter randomized controlled trials. J Thromb Thrombolysis 2022; 54:420-430. [PMID: 35922578 PMCID: PMC9362611 DOI: 10.1007/s11239-022-02681-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2022] [Indexed: 12/24/2022]
Abstract
Arterial and venous thrombotic events in COVID-19 cause significant morbidity and mortality among patients. Although international guidelines agree on the need for anticoagulation, it is unclear whether full-dose heparin anticoagulation confers additional benefits over prophylactic-dose anticoagulation. This systematic review and meta-analysis aimed to investigate the efficacy and safety of heparin full-dose anticoagulation in hospitalized non-critically ill COVID-19 patients. We searched Pubmed/Medline, EMBASE, Clinicaltrials.gov, medRxiv.org and Cochrane Central Register of clinical trials dated up to April 2022. Randomized controlled trials (RCTs) comparing full-dose heparin anticoagulation to prophylactic-dose anticoagulation or standard treatment in hospitalized non-critically ill COVID-19 patients were included in our pooled analysis. The primary endpoint was the rate of major thrombotic events and the co-primary endpoint was the rate of major bleeding events. We identified 4 studies, all of them multicenter, randomizing 2926 patients. Major thrombotic events were 23/1524 (1.5%) in full-dose heparin anticoagulation versus 57/1402 (4.0%) in prophylactic-dose [relative risk (RR) 0.39; 95% confidence interval (CI) 0.25–0.62; p˂0.01; I2 = 0%]. Clinical relevant bleeding events occurred in 1.7% (26/1524) among patients treated with heparin full anticoagulation dose compared to 1.1% (15/1403) in prophylactic-dose group (RR 1.60; 95% CI 0.85–3.03; p = 0.15; I2 = 20%). Mortality was 6.6% (101/1524) versus 8.6% (121/1402) (RR 0.63; 95% CI 0.33–1.19; p = 0.15). In this meta-analysis of high quality multicenter randomized trials, full-dose anticoagulation with heparin was associated with lower rate of major thrombotic events without differences in bleeding risk and mortality in hospitalized non critically ill COVID-19 patients. Study registration PROSPERO, review no. CRD42022301874.
Collapse
Affiliation(s)
- Eros Pilia
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy.,Department of Anesthesia, Resuscitation and Pain Therapy, University of Cagliari, Cagliari, Italy
| | - Alessandro Belletti
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Stefano Fresilli
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Gabriele Finco
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy.,Department of Anesthesia, Resuscitation and Pain Therapy, University of Cagliari, Cagliari, Italy
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy. .,School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
134
|
Nab L, Groenwold RHH, Klok FA, Bhoelan S, Kruip MJHA, Cannegieter SC. Estimating incidence of venous thromboembolism in COVID-19: Methodological considerations. Res Pract Thromb Haemost 2022; 6:e12776. [PMID: 35992195 PMCID: PMC9376932 DOI: 10.1002/rth2.12776] [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: 03/04/2022] [Revised: 06/24/2022] [Accepted: 07/05/2022] [Indexed: 12/15/2022] Open
Abstract
Background Coagulation abnormalities and coagulopathy are recognized as consequences of severe acute respiratory syndrome coronavirus 2 infection and the resulting coronavirus disease 2019 (COVID-19). Specifically, venous thromboembolism (VTE) has been reported as a frequent complication. By May 27, 2021, at least 93 original studies and 25 meta-analyses investigating VTE incidence in patients with COVID-19 had been published, showing large heterogeneity in reported VTE incidence ranging from 0% to 85%. This large variation complicates interpretation of individual study results as well as comparisons across studies, for example, to investigate changes in incidence over time, compare subgroups, and perform meta-analyses. Objectives This study sets out to provide an overview of sources of heterogeneity in VTE incidence studies in patients with COVID-19, illustrated using examples. Methods The original studies of three meta-analyses were screened and a list of sources of heterogeneity that may explain observed heterogeneity across studies was composed. Results The sources of heterogeneity in VTE incidence were classified as clinical sources and methodologic sources. Clinical sources of heterogeneity include differences between studies regarding patient characteristics that affect baseline VTE risk and protocols used for VTE testing. Methodologic sources of heterogeneity include differences in VTE inclusion types, data quality, and the methods used for data analysis. Conclusions To appreciate reported estimates of VTE incidence in patients with COVID-19 in relation to its etiology, prevention, and treatment, researchers should unambiguously report about possible clinical and methodological sources of heterogeneity in those estimates. This article provides suggestions for that.
Collapse
Affiliation(s)
- Linda Nab
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Rolf H. H. Groenwold
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Biomedical Data SciencesLeiden University Medical CenterLeidenThe Netherlands
| | - Frederikus A. Klok
- Department of Medicine – Thrombosis and HaemostatisLeiden University Medical CenterLeidenThe Netherlands
| | - Soerajja Bhoelan
- Department of HaematologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | | | - Suzanne C. Cannegieter
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Medicine – Thrombosis and HaemostatisLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
135
|
Chi G, Lee JJ, Memar Montazerin S, Marszalek J. Association of D-dimer with short-term risk of venous thromboembolism in acutely ill medical patients: A systematic review and meta-analysis. Vasc Med 2022; 27:478-486. [PMID: 35913041 DOI: 10.1177/1358863x221109855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND D-dimer, a marker of ongoing procoagulant activity, has been widely used for the diagnosis of venous thromboembolism (VTE). The prognostic significance of D-dimer in stratifying VTE risk for acutely ill medical patients has not been well-established. METHODS A literature search was performed to collect studies that compared the incidence of short-term VTE between acutely ill medical patients with elevated or nonelevated D-dimer levels. The cutoff of D-dimer was 0.5 μg/mL or otherwise defined by included studies. The study endpoint was any occurrence of VTE (inclusive of deep vein thrombosis [DVT], pulmonary embolism, or VTE-related death) within 90 days of hospital presentation. A meta-analytic approach was employed to estimate the odds ratio (OR) with 95% CI by fitting random-effects models using the generic inverse variance weighted approach. RESULTS A total of 10 studies representing 31,119 acutely ill medical patients were included. Compared to those with nonelevated D-dimer levels, patients with elevated D-dimer had approximately threefold greater odds for short-term VTE within 90 days (OR, 3.28; 95% CI, 2.44 to 4.40; p < 0.0001). The association of elevated D-dimer with VTE composite (OR, 3.33; 95% CI, 2.20 to 5.02) and with DVT (OR, 3.26; 95% CI, 2.32 to 4.58) was comparable. The association was significant among patients who presented various acute medical illness (OR, 2.68; 95% CI, 2.01 to 3.58) and those who presented with acute stroke (OR, 3.25; 95% CI, 2.31 to 4.58). CONCLUSION Elevation of D-dimer was predictive of the occurrence of VTE within 90 days among acutely ill medical patients. PROSPERO Registration ID: CRD42021264555.
Collapse
Affiliation(s)
- Gerald Chi
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jane J Lee
- Baim Institute for Clinical Research, Boston, MA, USA
| | | | - Jolanta Marszalek
- David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
136
|
Burn E, Duarte-Salles T, Fernandez-Bertolin S, Reyes C, Kostka K, Delmestri A, Rijnbeek P, Verhamme K, Prieto-Alhambra D. Venous or arterial thrombosis and deaths among COVID-19 cases: a European network cohort study. THE LANCET INFECTIOUS DISEASES 2022; 22:1142-1152. [PMID: 35576963 PMCID: PMC9106320 DOI: 10.1016/s1473-3099(22)00223-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/11/2022]
Abstract
Background There are few data on the incidence of thrombosis among COVID-19 cases, with most research concentrated on hospitalised patients. We aimed to estimate the incidence of venous thromboembolism, arterial thromboembolism, and death among COVID-19 cases and to assess the impact of these events on the risks of hospitalisation and death. Methods We conducted a distributed network cohort study using primary care records from the Netherlands, Italy, Spain, and the UK, and outpatient specialist records from Germany. The Spanish database was linked to hospital admissions. Participants were followed up from the date of a diagnosis of COVID-19 or positive RT-PCR test for SARS-CoV-2 (index date) for 90 days. The primary study outcomes were venous thromboembolic events, arterial thromboembolic events, and death, all over the 90 days from the index date. We estimated cumulative incidences for the study outcomes. Multistate models were used to calculate adjusted hazard ratios (HRs) for the association between venous thromboembolism or arterial thromboembolism occurrence and risks of hospitalisation or COVID-19 fatality. Findings Overall, 909 473 COVID-19 cases and 32 329 patients hospitalised with COVID-19 on or after Sept 1, 2020, were studied. The latest index dates across the databases ranged from Jan 30, 2021, to July 31, 2021. Cumulative 90-day incidence of venous thromboembolism ranged from 0·2% to 0·8% among COVID-19 cases, and up to 4·5% for those hospitalised. For arterial thromboembolism, estimates ranged from 0·1% to 0·8% among COVID-19 cases, increasing to 3·1% among those hospitalised. Case fatality ranged from 1·1% to 2·0% among patients with COVID-19, rising to 14·6% for hospitalised patients. The occurrence of venous thromboembolism in patients with COVID-19 was associated with an increased risk of death (adjusted HRs 4·42 [3·07–6·36] for those not hospitalised and 1·63 [1·39–1·90] for those hospitalised), as was the occurrence of arterial thromboembolism (3·16 [2·65–3·75] and 1·93 [1·57–2·37]). Interpretation Risks of venous thromboembolism and arterial thromboembolism were up to 1% among COVID-19 cases, and increased with age, among males, and in those who were hospitalised. Their occurrence was associated with excess mortality, underlying the importance of developing effective treatment strategies that reduce their frequency. Funding European Medicines Agency.
Collapse
Affiliation(s)
- Edward Burn
- Fundació Institut Universitari per a la Recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Barcelona, Spain; Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Talita Duarte-Salles
- Fundació Institut Universitari per a la Recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Barcelona, Spain
| | - Sergio Fernandez-Bertolin
- Fundació Institut Universitari per a la Recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Barcelona, Spain
| | - Carlen Reyes
- Fundació Institut Universitari per a la Recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Barcelona, Spain
| | - Kristin Kostka
- Real World Solutions, IQVIA, Cambridge, MA, USA; The Observational Health Data Sciences and Informatics Center at The Roux Institute, Northeastern University, Portland, ME, USA
| | - Antonella Delmestri
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Peter Rijnbeek
- Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Katia Verhamme
- Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Daniel Prieto-Alhambra
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK; Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
137
|
LaVasseur C, Neukam S, Kartika T, Samuelson Bannow B, Shatzel J, DeLoughery TG. Hormonal therapies and venous thrombosis: Considerations for prevention and management. Res Pract Thromb Haemost 2022; 6:e12763. [PMID: 36032216 PMCID: PMC9399360 DOI: 10.1002/rth2.12763] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 04/12/2022] [Accepted: 05/21/2022] [Indexed: 11/18/2022] Open
Abstract
Background Venous thromboses are well-established complications of hormonal therapy. Thrombosis risk is seen with both hormonal contraceptive agents and with hormone replacement therapy for menopause and gender transition. Over the past several decades, large epidemiological studies have helped better define these risks. Objectives To review and discuss the differences in thrombosis risk of the many of hormonal preparations available as well as their interaction with patient-specific factors. Methods We conducted a narrative review of the available literature regarding venous thrombosis and hormonal therapies including for contraception, menopausal symptoms, and gender transition. Results Thrombosis risk with estrogen-containing compounds increases with increasing systemic dose of estrogen. While progesterone-only-containing products are not associated with thrombosis, when paired with estrogen in combined oral contraceptives, the formulation of progesterone does impact the risk. These components, along with patient-specific factors, may influence the choice of hormonal preparation. For patients who develop thrombosis on hormonal treatment, anticoagulation is protective against future thrombosis. Duration of anticoagulation is dependent on ongoing and future hormone therapy choice. Finally, the optimal management of hormone therapy for individuals diagnosed with prothrombotic illnesses such as COVID-19 remains unclear. Conclusions When contemplating hormonal contraception or hormone replacement therapy, clinicians must consider a variety of factors including hormone type, dose, route, personal and family history of thrombosis, and other prothrombotic risk factors to make informed, personalized decisions regarding the risk of venous thrombosis.
Collapse
Affiliation(s)
- Corinne LaVasseur
- Department of MedicineOregon Health and Sciences UniversityPortlandOregonUSA
| | - Suvi Neukam
- Department of MedicineOregon Health and Sciences UniversityPortlandOregonUSA
- Division of Internal MedicineOregon Health and Sciences UniversityPortlandOregonUSA
| | - Thomas Kartika
- Division of Hematology‐OncologyOregon Health and Sciences UniversityPortlandOregonUSA
| | - Bethany Samuelson Bannow
- Department of MedicineOregon Health and Sciences UniversityPortlandOregonUSA
- Division of Hematology‐OncologyOregon Health and Sciences UniversityPortlandOregonUSA
- The Hemophilia CenterOregon Health and Sciences UniversityPortlandOregonUSA
| | - Joseph Shatzel
- Department of MedicineOregon Health and Sciences UniversityPortlandOregonUSA
- Division of Hematology‐OncologyOregon Health and Sciences UniversityPortlandOregonUSA
| | - Thomas G. DeLoughery
- Department of MedicineOregon Health and Sciences UniversityPortlandOregonUSA
- Division of Hematology‐OncologyOregon Health and Sciences UniversityPortlandOregonUSA
- Division of Laboratory Medicine, Department of PathologyOregon Health and Sciences UniversityPortlandOregonUSA
| |
Collapse
|
138
|
Abstract
COVID-19 is a primary respiratory illness that is frequently complicated by systemic involvement of the vasculature. Vascular involvement leads to an array of complications ranging from thrombosis to pulmonary edema secondary to loss of barrier function. This review will address the vasculopathy of COVID-19 with a focus on the role of the endothelium in orchestrating the systemic response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The endothelial receptor systems and molecular pathways activated in the setting of COVID-19 and the consequences of these inflammatory and prothrombotic changes on endothelial cell function will be discussed. The sequelae of COVID-19 vascular involvement at the level of organ systems will also be addressed, with an emphasis on the pulmonary vasculature but with consideration of effects on other vascular beds. The dramatic changes in endothelial phenotypes associated with COVID-19 has enabled the identification of biomarkers that could help guide therapy and predict outcomes. Knowledge of vascular pathogenesis in COVID-19 has also informed therapeutic approaches that may control its systemic sequelae. Because our understanding of vascular response in COVID-19 continues to evolve, we will consider areas of controversy, such as the extent to which SARS-CoV-2 directly infects endothelium and the degree to which vascular responses to SARS-CoV-2 are unique or common to those of other viruses capable of causing severe respiratory disease. This conceptual framework describing how SARS-CoV-2 infection affects endothelial inflammation, prothrombotic transformation, and barrier dysfunction will provide a context for interpreting new information as it arises addressing the vascular complications of COVID-19.
Collapse
Affiliation(s)
| | | | - Alec A Schmaier
- Division of Hemostasis and Thrombosis and
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
139
|
Thomas MR, Scully M. Clinical features of thrombosis and bleeding in COVID-19. Blood 2022; 140:184-195. [PMID: 35452509 PMCID: PMC9040438 DOI: 10.1182/blood.2021012247] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 04/13/2022] [Indexed: 01/08/2023] Open
Abstract
Infection with the SARS-CoV-2 virus, resulting in COVID-19 disease, has presented a unique scenario associated with high rates of thrombosis. The risk of venous thrombosis is some three- to sixfold higher than for patients admitted to a hospital for other indications, and for patients who have thrombosis, mortality appears to increase. Thrombosis may be a presenting feature of COVID-19. Pulmonary thrombi are the most frequent events, some related to deep vein thrombosis, but also to in situ microvascular and macrovascular thrombosis. Other venous thromboses include catheter- and circuit-associated in patients requiring hemofiltration and extracorporeal membrane oxygenation. Arterial thrombosis is less commonly documented, with 3% of patients in intensive care units having major arterial strokes and up to 9% having myocardial infarction, both of which are most likely multifactorial. Risk factors for thrombosis above those already documented in hospital settings include duration of COVID-19 symptoms before admission to the hospital. Laboratory parameters associated with higher risk of thrombosis include higher D-dimer, low fibrinogen, and low lymphocyte count, with higher factor VIII and von Willebrand factor levels indicative of more severe COVID-19 infection. All patients should receive thromboprophylaxis when admitted with COVID-19 infection, but the dose and length of treatment are still debated. Thrombosis continues to be treated according to standard VTE guidelines, but adjustments may be needed depending on other factors relevant to the patient's admission.
Collapse
Affiliation(s)
- Mari R Thomas
- Department of Haematology, University College London Hospital (UCLH), London, United Kingdom; and
- Cardiometabolic Programme, National Institute for Health and Care Research (NIHR), UCLH, University College London Biomedical Research Centre (BRC), London, United Kingdom
| | - Marie Scully
- Department of Haematology, University College London Hospital (UCLH), London, United Kingdom; and
- Cardiometabolic Programme, National Institute for Health and Care Research (NIHR), UCLH, University College London Biomedical Research Centre (BRC), London, United Kingdom
| |
Collapse
|
140
|
Gianni P, Goldin M, Ngu S, Zafeiropoulos S, Geropoulos G, Giannis D. Complement-mediated microvascular injury and thrombosis in the pathogenesis of severe COVID-19: A review. World J Exp Med 2022; 12:53-67. [PMID: 36157337 PMCID: PMC9350720 DOI: 10.5493/wjem.v12.i4.53] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/27/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) causes acute microvascular thrombosis in both venous and arterial structures which is highly associated with increased mortality. The mechanisms leading to thromboembolism are still under investigation. Current evidence suggests that excessive complement activation with severe amplification of the inflammatory response (cytokine storm) hastens disease progression and initiates complement-dependent cytotoxic tissue damage with resultant prothrombotic complications. The concept of thromboinflammation, involving overt inflammation and activation of the coagulation cascade causing thrombotic microangiopathy and end-organ damage, has emerged as one of the core components of COVID-19 pathogenesis. The complement system is a major mediator of the innate immune response and inflammation and thus an appealing treatment target. In this review, we discuss the role of complement in the development of thrombotic microangiopathy and summarize the current data on complement inhibitors as COVID-19 therapeutics.
Collapse
Affiliation(s)
- Panagiota Gianni
- Department of Internal Medicine III, Hematology, Oncology, Palliative Medicine, Rheumatology and Infectious Diseases, University Hospital Ulm, Ulm 89070, Germany
| | - Mark Goldin
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, New York, NY 11549, United States
- Feinstein Institutes for Medical Research at Northwell Health, Feinstein Institutes , New York, NY 11030, United States
| | - Sam Ngu
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, New York, NY 11549, United States
| | - Stefanos Zafeiropoulos
- Elmezzi Graduate School of Molecular Medicine, Northwell Health, New York, NY 11030, United States
| | - Georgios Geropoulos
- Department of General Surgery, University College London Hospitals, London NW12BU, United Kingdom
| | - Dimitrios Giannis
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, New York, NY 11549, United States
- North Shore/Long Island Jewish General Surgery, Northwell Health, New York, NY 11021, United States
| |
Collapse
|
141
|
Johnson SE, Pai E, Voroba A, Chen NW, Bahl A. Examining D-dimer and Empiric Anti-coagulation in COVID-19-Related Thrombosis. Cureus 2022; 14:e26883. [PMID: 35978762 PMCID: PMC9375952 DOI: 10.7759/cureus.26883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Thrombosis is thought to occur frequently in the setting of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We aimed to elucidate the relationship between macro/microvascular thrombosis, D-dimer levels, and empiric anticoagulation in coronavirus disease 2019 (COVID-19). Methods This was an exploratory prospective, single-site, observational study. Adult emergency department patients with COVID-19 requiring hospitalization received a point-of-care lower extremity venous duplex ultrasound. The primary endpoint was thromboembolism and associated D-dimer level. Secondary endpoints included rates of micro and macro thrombotic complications as well as empiric anticoagulant use. Results Between January 13th and April 12th 2021, 52 patients were enrolled. Median D-dimer at presentation was 650 ng/mL (range 250-10,000 ng/mL) among patients with negative duplex studies. During hospitalization, 18 patients underwent 20 additional studies assessing for venous thromboembolism (VTE). These studies yielded one deep vein thrombosis (DVT) diagnosis. Among patients with negative studies median D-dimer was 1,246 ng/mL (range 329-10,000 ng/mL). Two patients experienced microvascular complications. Seven patients were started on empiric full dose anticoagulation. Conclusion While VTE remains a major concern amongst patients with COVID-19, the normal D-dimer cut off of >500 ng/mL likely should not be used to initiate further VTE workup. Additionally, moderately elevated D-dimer did not correlate strongly with microvascular complications and may not be relevant in the decision to initiate empiric anticoagulation.
Collapse
|
142
|
Krutzke L, Rösler R, Allmendinger E, Engler T, Wiese S, Kochanek S. Process- and product-related impurities in the ChAdOx1 nCov-19 vaccine. eLife 2022; 11:78513. [PMID: 35781137 PMCID: PMC9313527 DOI: 10.7554/elife.78513] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/03/2022] [Indexed: 11/30/2022] Open
Abstract
ChAdOx1 nCov-19 and Ad26.COV2.S are approved vaccines inducing protective immunity against SARS-CoV-2 infection in humans by expressing the Spike protein of SARS-CoV-2. We analyzed protein content and protein composition of ChAdOx1 nCov-19 and Ad26.COV2.S by biochemical methods and by mass spectrometry. Four out of four tested lots of ChAdOx1 nCoV-19 contained significantly higher than expected levels of host cell proteins (HCPs) and of free viral proteins. The most abundant contaminating HCPs belonged to the heat-shock protein and cytoskeletal protein families. The HCP content exceeded the 400 ng specification limit per vaccine dose, as set by the European Medicines Agency (EMA) for this vaccine, by at least 25-fold and the manufacturer’s batch-release data in some of the lots by several hundred-fold. In contrast, three tested lots of the Ad26.COV2.S vaccine contained only very low amounts of HCPs. As shown for Ad26.COV2.S production of clinical grade adenovirus vaccines of high purity is feasible at an industrial scale. Correspondingly, purification procedures of the ChAdOx1 nCov-19 vaccine should be modified to remove protein impurities as good as possible. Our data also indicate that standard quality assays, as they are used in the manufacturing of proteins, have to be adapted for vectored vaccines.
Collapse
Affiliation(s)
- Lea Krutzke
- Department of Gene Therapy, University of Ulm, Ulm, Germany
| | - Reinhild Rösler
- Core Unit Mass Spectrometry and Proteomics, University of Ulm, Ulm, Germany
| | | | - Tatjana Engler
- Department of Gene Therapy, University of Ulm, Ulm, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, University of Ulm, Ulm, Germany
| | | |
Collapse
|
143
|
Spyropoulos AC. Extended post-discharge thromboprophylaxis in hospitalized COVID-19 patients. Expert Rev Hematol 2022; 15:597-605. [PMID: 35786377 DOI: 10.1080/17474086.2022.2098104] [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: 11/04/2022]
Abstract
INTRODUCTION Hospitalized COVID-19 patients, particularly those with high risk features, are at risk for venous and arterial thromboembolic events for approximately 30 days or more after hospital discharge. Extended post-hospital discharge thromboprophylaxis has potential to reduce this risk. AREAS COVERED Recent cohort, registry, and randomized trial data on the topic of extended post-discharge thromboprophylaxis in COVID-19 inpatients are reviewed, and key patient subgroups at high thrombotic risk are highlighted, with antithrombotic guidelines on the topic discussed. EXPERT OPINION COVID-19 inpatients with cardiovascular risk factors, advanced age, intensive care unit stay, or an IMPROVE VTE score of 4 or more or a score of 2 or 3 plus elevated D-dimers (> twice the upper limit of normal) or an IMPROVE-DD VTE score of ≥ 4 are at high thrombotic risk in the post-discharge period. These high risk patient subgroups benefit from extended post-discharge thromboprophylaxis, specifically with rivaroxaban 10mg daily for 35 days. Recent NIH and ISTH guidelines recommend or suggest this approach. Results from other clinical trials are pending.
Collapse
Affiliation(s)
- Alex C Spyropoulos
- Institute of Health Systems Science - Feinstein Institutes for Medical Research and The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, and Department of Medicine, Anticoagulation and Clinical Thrombosis Services, Northwell Health at Lenox Hill Hospital, New York, NY, USA
| |
Collapse
|
144
|
Ferreira-da-Silva R, Ribeiro-Vaz I, Morato M, Junqueira Polónia J. A comprehensive review of adverse events to drugs used in COVID-19 patients: Recent clinical evidence. Eur J Clin Invest 2022; 52:e13763. [PMID: 35224719 PMCID: PMC9111855 DOI: 10.1111/eci.13763] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/16/2022] [Accepted: 02/20/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND Since the breakthrough of the pandemic, several drugs have been used to treat COVID-19 patients. This review aims to gather information on adverse events (AE) related to most drugs used in this context. METHODS We performed a literature search to find articles that contained information about AE in COVID-19 patients. We analysed and reviewed the most relevant studies in the Medline (via PubMed), Scopus and Web of Science. The most frequent AE identified were grouped in our qualitative analysis by System Organ Class (SOC), the highest level of the MedDRA medical terminology for each of the drugs studied. RESULTS The most frequent SOCs among the included drugs are investigations (n = 7 drugs); skin and subcutaneous tissue disorders (n = 5 drugs); and nervous system disorders, infections and infestations, gastrointestinal disorders, hepatobiliary disorders, and metabolism and nutrition disorders (n = 4 drugs). Other SOCs also emerged, such as general disorders and administration site conditions, renal and urinary disorders, vascular disorders and cardiac disorders (n = 3 drugs). Less frequent SOC were eye disorders, respiratory, thoracic and mediastinal disorders, musculoskeletal and connective tissue disorders, and immune system disorders (n = 2 drugs). Psychiatric disorders, and injury, poisoning and procedural complications were also reported (n = 1 drug). CONCLUSIONS Some SOCs seem to be more frequent than others among the COVID-19 drugs included, although neither of the studies included reported causality analysis. For that purpose, further clinical studies with robust methodologies, as randomised controlled trials, should be designed and performed.
Collapse
Affiliation(s)
- Renato Ferreira-da-Silva
- Porto Pharmacovigilance Centre, INFARMED, I.P, University of Porto, Porto, Portugal.,Department of Community Medicine, Health Information and Decision, University of Porto, Porto, Portugal.,CINTESIS - Center for Health Technology and Services Research, Porto, Portugal
| | - Inês Ribeiro-Vaz
- Porto Pharmacovigilance Centre, INFARMED, I.P, University of Porto, Porto, Portugal.,Department of Community Medicine, Health Information and Decision, University of Porto, Porto, Portugal.,CINTESIS - Center for Health Technology and Services Research, Porto, Portugal
| | - Manuela Morato
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, University of Porto, Porto, Portugal
| | - Jorge Junqueira Polónia
- Porto Pharmacovigilance Centre, INFARMED, I.P, University of Porto, Porto, Portugal.,CINTESIS - Center for Health Technology and Services Research, Porto, Portugal.,Department of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
145
|
Andersen KM, Joseph CS, Mehta HB, Streiff MB, Betz JF, Bollinger RC, Fisher AM, Gupta A, LeMaistre CF, Robinson ML, Xu Y, Ng DK, Alexander GC, Garibaldi BT. Thromboprophylaxis in people hospitalized with COVID-19: Assessing intermediate or standard doses in a retrospective cohort study. Res Pract Thromb Haemost 2022; 6:e12753. [PMID: 35859579 PMCID: PMC9287673 DOI: 10.1002/rth2.12753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/09/2022] [Accepted: 05/27/2022] [Indexed: 11/08/2022] Open
Abstract
Background and Objectives Current clinical guidelines recommend thromboprophylaxis for adults hospitalized with coronavirus disease 2019 (COVID-19), yet it is unknown whether higher doses of thromboprophylaxis offer benefits beyond standard doses. Methods We studied electronic health records from 50 091 adults hospitalized with COVID-19 in the United States between February 2020 and February 2021. We compared standard (enoxaparin 30 or 40 mg/day, fondaparinux 2.5 mg, or heparin 5000 units twice or thrice per day) versus intermediate (enoxaparin 30 or 40 mg twice daily, or up to 1.2 mg/kg of body weight daily, heparin 7500 units thrice per day or heparin 10 000 units twice or thrice per day) thromboprophylaxis. We separately examined risk of escalation to therapeutic anticoagulation, severe disease (first occurrence of high-flow nasal cannula, noninvasive positive pressure ventilation or invasive mechanical ventilation), and death. To summarize risk, we present hazard ratios (HRs) with 95% confidence intervals (CIs) using adjusted time-dependent Cox proportional hazards regression models. Results People whose first dose was high intensity were younger, more often obese, and had greater oxygen support requirements. Intermediate dose thromboprophylaxis was associated with increased risk of therapeutic anticoagulation (HR, 3.39; 95% CI, 3.22-3.57), severe disease (HR, 1.22; 95% CI, 1.17-1.28), and death (HR, 1.37; 95% CI, 1.21-1.55). Increased risks associated with intermediate-dose thromboprophylaxis persisted in subgroup and sensitivity analyses varying populations and definitions of exposures, outcomes, and covariates. Conclusions Our findings do not support routine use of intermediate-dose thromboprophylaxis to prevent clinical worsening, severe disease, or death among adults hospitalized with COVID-19.
Collapse
Affiliation(s)
- Kathleen M. Andersen
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
- Center for Drug Safety and EffectivenessJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Corey S. Joseph
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
- Center for Drug Safety and EffectivenessJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Hemalkumar B. Mehta
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
- Center for Drug Safety and EffectivenessJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Michael B. Streiff
- Division of HematologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Joshua F. Betz
- Department of BiostatisticsJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Robert C. Bollinger
- Division of Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Center for Clinical Global Health EducationJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Arielle M. Fisher
- Sarah Cannon, Genospace, HCA Healthcare Research InstituteNashvilleTennesseeUSA
| | - Amita Gupta
- Division of Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of International HealthJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | | | - Matthew L. Robinson
- Division of Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Yanxun Xu
- Department of Applied Mathematics and StatisticsJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Derek K. Ng
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - G. Caleb Alexander
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
- Center for Drug Safety and EffectivenessJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
- Division of General Internal MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Brian T. Garibaldi
- Division of Pulmonary and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
146
|
Sochet AA, Morrison JM, Jaffray J, Godiwala N, Wilson HP, Thornburg CD, Bhat RV, Zia A, Lawrence C, Kudchadkar SR, Hamblin F, Russell CJ, Streiff MB, Spyropoulos AC, Amankwah EK, Goldenberg NA. Enoxaparin Thromboprophylaxis in Children Hospitalized for COVID-19: A Phase 2 Trial. Pediatrics 2022; 150:e2022056726. [PMID: 35484817 DOI: 10.1542/peds.2022-056726] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Evidence regarding the safety and efficacy of anticoagulant thromboprophylaxis among pediatric patients hospitalized for coronavirus disease 2019 (COVID-19) is limited. We sought to evaluate safety, dose-finding, and preliminary efficacy of twice-daily enoxaparin as primary thromboprophylaxis among children hospitalized for symptomatic COVID-19, including primary respiratory infection and multisystem inflammatory syndrome in children (MISC). METHODS We performed a phase 2, multicenter, prospective, open-label, single-arm clinical trial of twice-daily enoxaparin (initial dose: 0.5mg/kg per dose; max: 60mg; target anti-Xa activity: 0.20-0.49IU/mL) as primary thromboprophylaxis for children <18 years of age hospitalized for symptomatic COVID-19. Study endpoints included: cumulative incidence of International Society of Thrombosis and Haemostasis-defined clinically relevant bleeding; enoxaparin dose-requirements; and cumulative incidence of venous thromboembolism within 30-days of hospital discharge. Descriptive statistics summarized endpoint estimates that were further evaluated by participant age (±12 years) and clinical presentation. RESULTS Forty children were enrolled and 38 met analyses criteria. None experienced clinically relevant bleeding. Median (interquartile range) dose to achieve target anti-Xa levels was 0.5 mg/kg (0.48-0.54). Dose-requirement did not differ by age (0.5 [0.46-0.52] mg/kg for age ≥12 years versus 0.52 [0.49-0.55] mg/kg for age <12 years, P = .51) but was greater for participants with MISC (0.52 [0.5-0.61] mg/kg) as compared with primary COVID-19 (0.48 [0.39-0.51] mg/kg, P = .010). Two children (5.3%) developed central-venous catheter-related venous thromboembolism. No serious adverse events were related to trial intervention. CONCLUSIONS Among children hospitalized for COVID-19, thromboprophylaxis with twice-daily enoxaparin appears safe and warrants further investigation to assess efficacy.
Collapse
Affiliation(s)
| | | | - Julie Jaffray
- Department of Pediatrics, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Nihal Godiwala
- Department of Pediatrics, Louisiana State University School of Medicine, New Orleans, Louisiana
| | - Hope P Wilson
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham, Alabama
| | - Courtney D Thornburg
- Department of Pediatrics, University of California San Diego, La Jolla, California Division of Hematology and Oncology, Rady Children's Hospital San Diego, San Diego, California
| | - Rukhmi V Bhat
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ayesha Zia
- Department of Pediatrics, The University of Texas Southwestern, Dallas, Texas
| | | | - Sapna R Kudchadkar
- Critical Care Medicine
- Pediatrics
- Physical Medicine and Rehabilitation, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Christopher J Russell
- Department of Pediatrics, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - Alex C Spyropoulos
- Department of Medicine, Zucker School of Medicine at Hofstra and Northwell and Feinstein Institutes for Medical Research, Manhasset, New York
- Department of Obstetrics and Gynecology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ernest K Amankwah
- Departments of Anesthesia
- Pediatrics
- Oncology, Johns Hopkins All Children's Institute for Clinical and Translational Research
| | | |
Collapse
|
147
|
Tandayam A, Syed MHN, Kumar G. Pathophysiology and Management of Hypercoagulation in Infectious Diseases. JOURNAL OF CARDIAC CRITICAL CARE TSS 2022. [DOI: 10.1055/s-0042-1757370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AbstractNumerous systemic infections may have hypercoagulation as one of the complications, which may range from asymptomatic presentation of elevation of biochemical markers of coagulation such as that of fibrin and thrombin generation, to a much severe, symptomatic, life-threatening, disseminated intravascular coagulation (DIC), which results in the formation of thrombi in the microvasculature of various organs. This phenomenon contributes to increase in morbidity and mortality in various infectious diseases. The current review discusses various mechanisms of hypercoagulation during infections such as tissue factor activation, endothelial cell activation, inhibition of physiological anticoagulant pathways, and fibrinolysis inhibition. The review also discusses pathophysiological changes in the coagulation system and its management in the recent pandemic of COVID-19. The article also discusses role of various parenteral and oral anticoagulants in the management of infectious diseases. The review provides clinical data on various anticoagulants used during hospitalization and extended prophylaxis for the management of venous thromboembolism in various infections.
Methodology Because this is a review of published literature and no humans or animals were involved, ethical committee approval was not required and patient consent was not required.
Collapse
Affiliation(s)
- Abhishek Tandayam
- Medical Affairs Department, Dr Reddy's Laboratories, Hyderabad, India
| | | | - Gaurav Kumar
- Medical Affairs Department, Dr Reddy's Laboratories, Hyderabad, India
| |
Collapse
|
148
|
Woller SC, Stevens SM, Bledsoe JR, Fazili M, Lloyd JF, Snow GL, Horne BD. Biomarker derived risk scores predict venous thromboembolism and major bleeding among patients with COVID-19. Res Pract Thromb Haemost 2022; 6:e12765. [PMID: 35873221 PMCID: PMC9301476 DOI: 10.1002/rth2.12765] [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: 11/30/2021] [Revised: 05/21/2022] [Accepted: 06/19/2022] [Indexed: 12/15/2022] Open
Abstract
Background Venous thromboembolism (VTE) risk is increased in patients with COVID-19 infection. Understanding which patients are likely to develop VTE may inform pharmacologic VTE prophylaxis decision making. The hospital-associated venous thromboembolism-Intermountain Risk Score (HA-VTE IMRS) and the hospital-associated major bleeding-Intermountain Risk Score (HA-MB IMRS) are risk scores predictive of VTE and bleeding that were derived from only patient age and data found in the complete blood count (CBC) and basic metabolic panel (BMP). Objectives We assessed the HA-VTE IMRS and HA-MB IMRS for predictiveness of 90-day VTE and major bleeding, respectively, among patients diagnosed with COVID-19, and further investigated if adding D-dimer improved these predictions. We also reported 30-day outcomes. Patients/Methods We identified 5047 sequential patients with a laboratory confirmed diagnosis of COVID-19 and a CBC and BMP between 2 days before and 7 days following the diagnosis of COVID-19 from March 12, 2020, to February 28, 2021. We calculated the HA-VTE IMRS and the HA-MB IMRS for all patients. We assessed the added predictiveness of D-dimer obtained within 48 hours of the COVID test. Results The HA-VTE IMRS yielded a c-statistic of 0.70 for predicting 90-day VTE and adding D-dimer improved the c-statistic to 0.764 with the corollary sensitivity/specificity/positive/negative predictive values of 49.4%/75.7%/6.7%/97.7% and 58.8%/76.2%/10.9%/97.4%, respectively. Among hospitalized and ambulatory patients separately, the HA-VTE IMRS performed similarly. The HA-MB IMRS predictiveness for 90-day major bleeding yielded a c-statistic of 0.64. Conclusion The HA-VTE IMRS and HA-MB IMRS predict 90- and 30-day VTE and major bleeding among COVID-19 patients. Adding D-dimer improved the predictiveness of the HA-VTE IMRS for VTE.
Collapse
Affiliation(s)
- Scott C. Woller
- Department of MedicineIntermountain Medical Center, Intermountain HealthcareMurrayUtahUSA
- Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Scott M. Stevens
- Department of MedicineIntermountain Medical Center, Intermountain HealthcareMurrayUtahUSA
- Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Joseph R. Bledsoe
- Department of Emergency Medicine, Intermountain Medical CenterIntermountain HealthcareMurrayUtahUSA
- Stanford UniversityStanfordCaliforniaUSA
| | - Masarret Fazili
- Department of MedicineIntermountain Medical Center, Intermountain HealthcareMurrayUtahUSA
| | - James F. Lloyd
- Department of InformaticsIntermountain Medical Center, Intermountain HealthcareMurrayUtahUSA
| | - Greg L. Snow
- Intermountain Statistical Data Center, Intermountain Medical CenterIntermountain HealthcareMurrayUtahUSA
| | - Benjamin D. Horne
- Intermountain Medical Center Heart InstituteMurrayUtahUSA
- Division of Cardiovascular MedicineStanford UniversityStanfordCaliforniaUSA
| |
Collapse
|
149
|
Luijten D, de Jong CMM, Klok FA. Post Pulmonary Embolism Syndrome. Arch Bronconeumol 2022; 58:533-535. [PMID: 35312578 DOI: 10.1016/j.arbres.2021.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 11/19/2022]
Affiliation(s)
- Dieuwke Luijten
- Department of Medicine - Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Cindy M M de Jong
- Department of Medicine - Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Frederikus A Klok
- Department of Medicine - Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
150
|
Mansory EM, Abu-Farhaneh M, Iansavitchene A, Lazo-Langner A. Venous and Arterial Thrombosis in Ambulatory and Discharged COVID-19 Patients: A Systematic Review and Meta-analysis. TH OPEN 2022; 6:e276-e282. [PMID: 36299810 PMCID: PMC9484870 DOI: 10.1055/a-1913-4377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 07/25/2022] [Indexed: 11/12/2022] Open
Abstract
Introduction
Venous and arterial thromboses are frequently observed complications in patients with severe novel coronavirus disease 2019 (COVID-19) infection who require hospital admission. In this study, we evaluate the epidemiology of venous and arterial thrombosis events in ambulatory and postdischarge patients with COVID-19 infection.
Materials and Method
EMBASE and MEDLINE were searched up to July 21, 2021, in addition to other sources. We included studies that assessed the epidemiology of venous and arterial thrombosis events in ambulatory and postdischarge COVID-19 patients.
Results
A total of 16 studies (102,779 patients) were identified. The overall proportion of venous thromboembolic events in all patients, that is, ambulatory and postdischarge, was 0.80% (95% confidence interval [CI]: 0.44–1.28), 0.28% (95% CI: 0.07–0.64), and 1.16% (95% CI: 0.69–1.74), respectively. Arterial events occurred in 0.75% (95% CI: 0.27–1.47) of all patients, 1.45% (95% CI: 1.10–1.86) of postdischarge patients, and 0.23% (95% CI: 0.019–0.66) of ambulatory patients. The pooled incidence rate estimates per 1,000 patient-days for VTE events were 0.06 (95% CI: 0.03–0.08) and 0.12 (95% CI: 0.07–0.19) for outpatients and postdischarge, respectively, whereas for arterial events were 0.10 (95% CI: 0–0.30) and 0.26 (95% CI: 0.16–0.37).
Conclusion
This study found a low risk of venous and arterial thrombi in ambulatory and postdischarge COVID-19 patients, with a higher risk in postdischarge patients compared with ambulatory patients. This suggests that regular universal thromboprophylaxis in these patient populations is probably not necessary.
Collapse
Affiliation(s)
- Eman M. Mansory
- Department of Medicine, Division of Hematology, Western University, London, Ontario, Canada
- Department of Hematology, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Alla Iansavitchene
- Health Sciences Library, London Health Sciences Centre, London, Ontario, Canada
| | - Alejandro Lazo-Langner
- Department of Medicine, Division of Hematology, Western University, London, Ontario, Canada
- Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada
| |
Collapse
|