201
|
Gómez-Porro P, Cabal-Paz B, Valenzuela-Chamorro S, Desanvicente-Celis Z, Sabin-Muñoz J, Ochoa-López C, Flórez C, Enríquez-Calzada S, Martín-García R, Esain-González Í, García-Fleitas B, Silva-Hernández L, Ruiz-Molina Á, Gamo-González E, Durán-Lozano A, Velasco-Calvo R, Alba-Alcántara L, González-Santiago R, Callejas-Díaz A, Brea-Álvarez B, Salazar-Uribe JC, Escamilla-Crespo C, Carneado-Ruiz J. High frequency of endoluminal thrombus in patients with ischaemic stroke following AARS-CoV-2 infection. Neurologia 2024; 39:43-54. [PMID: 38065431 DOI: 10.1016/j.nrleng.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 04/06/2021] [Indexed: 12/22/2023] Open
Abstract
BACKGROUND Ischaemic stroke may be a major complication of SARS-CoV-2 infection. Studying and characterising the different aetiological subtypes, clinical characteristics, and functional outcomes may be valuable in guiding patient selection for optimal management and treatment. METHODS Data were collected retrospectively on consecutive patients with COVID-19 who developed acute focal brain ischaemia (between 1 March and 19 April 2020) at a tertiary university hospital in Madrid (Spain). RESULTS During the study period, 1594 patients were diagnosed with COVID-19. We found 22 patients with ischaemic stroke (1.38%), 6 of whom did not meet the inclusion criteria. The remaining 16 patients were included in the study (15 cases of ischaemic stroke and one case of transient ischaemic attack). Median baseline National Institutes of Health Stroke Scale score was 9 (interquartile range: 16), and mean (standard deviation) age was 73 years (12.8). Twelve patients (75%) were men. Mean time from COVID-19 symptom onset to stroke onset was 13 days. Large vessel occlusion was identified in 12 patients (75%). We detected elevated levels of D-dimer in 87.5% of patients and C-reactive protein in 81.2%. The main aetiology was atherothrombotic stroke (9 patients, 56.3%), with the predominant subtype being endoluminal thrombus (5 patients, 31.2%), involving the internal carotid artery in 4 cases and the aortic arch in one. The mortality rate in our series was 44% (7 of 16 patients). CONCLUSIONS In patients with COVID-19, the most frequent stroke aetiology was atherothrombosis, with a high proportion of endoluminal thrombus (31.2% of patients). Our clinical and laboratory data support COVID-19-associated coagulopathy as a relevant pathophysiological mechanism for ischaemic stroke in these patients.
Collapse
Affiliation(s)
- P Gómez-Porro
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - B Cabal-Paz
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - S Valenzuela-Chamorro
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Z Desanvicente-Celis
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - J Sabin-Muñoz
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - C Ochoa-López
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - C Flórez
- Universidad CES, Medellín, Colombia
| | - S Enríquez-Calzada
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - R Martín-García
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Í Esain-González
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - B García-Fleitas
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - L Silva-Hernández
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Á Ruiz-Molina
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - E Gamo-González
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - A Durán-Lozano
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - R Velasco-Calvo
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - L Alba-Alcántara
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - R González-Santiago
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - A Callejas-Díaz
- Servicio de Medicina Interna, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - B Brea-Álvarez
- Servicio de Radiología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | | | - C Escamilla-Crespo
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - J Carneado-Ruiz
- Servicio de Neurología, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain.
| |
Collapse
|
202
|
Elfert K, Malik M, Aboursheid T, Mohamed M, Elfert Y, Beran A, Jaber F, Elromisy E, Al-Taee A, Kahaleh M. Impact of COVID-19 infection on patients admitted with nonvariceal upper gastrointestinal bleeding: an analysis from the National Inpatient Sample. Proc AMIA Symp 2023; 37:36-41. [PMID: 38174004 PMCID: PMC10761015 DOI: 10.1080/08998280.2023.2260280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 09/07/2023] [Indexed: 01/05/2024] Open
Abstract
Background Nonvariceal upper gastrointestinal bleeding (NVUGIB) is a medical emergency that has significant morbidity and mortality. The available data about the impact of COVID-19 infection on mortality in patients with NVUGIB is limited. Methods We identified all hospitalizations with a principal diagnosis of NVUGIB in 2020. The baseline characteristics and clinical outcomes of patients with COVID-19 infection were compared to those without COVID-19 infection. Results NVUGIB patients with COVID-19 infection had higher mortality (5% vs 2%, P < 0.0001), a longer mean length of stay (6.85 vs 4.48 days, P < 0.0001), and a lower rate of esophagogastroduodenoscopy utilization (40% vs 51%, P < 0.0001) than those without COVID-19 infection. Multivariate logistic regression analysis showed that COVID-19 infection was associated with a higher mortality rate (odds ratio 2.2, 95% confidence interval, 1.4-3.4). Conclusions COVID-19 infection is an independent predictor of mortality in adults hospitalized with NVUGIB.
Collapse
Affiliation(s)
- Khaled Elfert
- Department of Internal Medicine, SBH Health System, New York, New York, USA
| | - Mushrin Malik
- Department of Internal Medicine, SBH Health System, New York, New York, USA
| | - Tarek Aboursheid
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, Illinois, USA
| | - Mouhand Mohamed
- Department of Internal Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Yomna Elfert
- Department of Pediatrics, UH Cleveland Medical Center, Cleveland, Ohio, USA
| | - Azizullah Beran
- Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, Indiana, USA
| | - Fouad Jaber
- Department of Internal Medicine, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Esraa Elromisy
- Tanta University Faculty of Medicine, Tanta, Gharbiyah, Egypt
| | - Ahmad Al-Taee
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Michel Kahaleh
- Division of Gastroenterology, Department of Medicine, Robert Wood Johnson University Hospital, New Brunswick, New Jersey, USA
| |
Collapse
|
203
|
Xiao S, Yuan Z, Huang Y. The Potential Role of Nitric Oxide as a Therapeutic Agent against SARS-CoV-2 Infection. Int J Mol Sci 2023; 24:17162. [PMID: 38138990 PMCID: PMC10742813 DOI: 10.3390/ijms242417162] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become the greatest worldwide public health threat of this century, which may predispose multi-organ failure (especially the lung) and death despite numerous mild and moderate symptoms. Recent studies have unraveled the molecular and clinical characteristics of the infectivity, pathogenicity, and immune evasion of SARS-CoV-2 and thus improved the development of many different therapeutic strategies to combat COVID-19, including treatment and prevention. Previous studies have indicated that nitric oxide (NO) is an antimicrobial and anti-inflammatory molecule with key roles in pulmonary vascular function in the context of viral infections and other pulmonary disease states. This review summarized the recent advances of the pathogenesis of SARS-CoV-2, and accordingly elaborated on the potential application of NO in the management of patients with COVID-19 through antiviral activities and anti-inflammatory properties, which mitigate the propagation of this disease. Although there are some limits of NO in the treatment of COVID-19, it might be a worthy candidate in the multiple stages of COVID-19 prevention or therapy.
Collapse
Affiliation(s)
| | | | - Yi Huang
- National Biosafety Laboratory, Chinese Academy of Sciences, Wuhan 430020, China
| |
Collapse
|
204
|
Ludhiadch A, Paul SR, Khan R, Munshi A. COVID-19 induced ischemic stroke and mechanisms of viral entry in brain and clot formation: a systematic review and current update. Int J Neurosci 2023; 133:1153-1166. [PMID: 35412938 DOI: 10.1080/00207454.2022.2056460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/16/2022] [Indexed: 10/18/2022]
Abstract
Background: Coronavirus disease 2019, caused by SARS-CoV-2 (SCV-2) was stated as a pandemic on March 11 2020 by World Health Organization (WHO), and since then, it has become a major health issue worldwide. It mainly attacks the respiratory system with various accompanying complications, including cardiac injury, renal failure, encephalitis and Stroke.Materials and Methods: The current systematic review has been compiled to summarize the available literature on SCV-2 induced ischemic Stroke and its subtypes. Further, the mechanisms by which the virus crosses the blood-brain barrier (BBB) to enter the brain have also been explored. The role of CRP and D-dimer as potent prognostic markers was also explored. The literature search was carried out comprehensively on Google scholar, PubMed, SCOP US, Embase and Cochrane databases by following guidelines.Results: All the studies were reviewed thoroughly by authors and disagreements were resolved by consensus and help of the senior authors. The most common subtype of the IS was found to be large artery atherosclerosis in SCV-2 induced IS. Hypertension emerged as the most significant risk factor. The mechanism resulting in elevated levels of CRP and D-dimer have also been discussed. However, there is a scarcity of definitive evidence on how SCV-2 enters the human brain. The available literature based on various studies demonstrated that SCV-2 enters through the nasopharyngeal tract via olfactory cells to olfactory neurons, astrocytes and via choroid plexus through endothelial cells. Further, disruption of gut-brain axis has been also discussed.Conclusion: Data available in the literature is not adequate to come to a conclusion. Therefore, there is a need to carry out further studies to delineate the possible association between SCV-2 induced IS.
Collapse
Affiliation(s)
- Abhilash Ludhiadch
- Department of Human Genetics and Molecular Medicine Central, University of Punjab Bathinda, Bathinda, Punjab, India
| | - Swaraj Ranjan Paul
- Department of Human Genetics and Molecular Medicine Central, University of Punjab Bathinda, Bathinda, Punjab, India
| | - Rahul Khan
- Department of Human Genetics and Molecular Medicine Central, University of Punjab Bathinda, Bathinda, Punjab, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine Central, University of Punjab Bathinda, Bathinda, Punjab, India
| |
Collapse
|
205
|
Siddique YA, Chaudhry R, Ahmad M, Sebai A, Sharma L, Hassouba M, Virk GS. The Trend of Arrhythmias in Patients With COVID-19: A Complication or Late Manifestation? Cureus 2023; 15:e50746. [PMID: 38239526 PMCID: PMC10794791 DOI: 10.7759/cureus.50746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 01/22/2024] Open
Abstract
Patients diagnosed with coronavirus disease (CVD) who experience cardiovascular complications or have pre-existing cardiovascular disease are at an increased risk of death. The primary heart-related consequences associated with COVID-19 encompass venous thromboembolism, shock, heart failure, arrhythmias, myocarditis, acute myocardial infarction, and acute cardiac damage. The coronavirus has the potential to induce cardiovascular complications or exacerbate pre-existing CVD through various mechanisms. These mechanisms include dysregulation of the renin-angiotensin-aldosterone system; direct viral toxicity; damage to endothelial cells; formation of blood clots and subsequent inflammation, a phenomenon known as thromboinflammation; an excessive immune response known as cytokine storm; and an imbalance between the demand and supply of oxygen in the body. In this study, we comprehensively analyze the cardiovascular symptoms, histology, and underlying mechanisms associated with COVID-19. Our aim is to contribute to the identification of future research objectives and aid in the advancement of therapeutic management approaches.
Collapse
Affiliation(s)
- Yusuf A Siddique
- Basic Sciences, St. George's University School of Medicine, True Blue, GRD
| | | | | | - Ahmad Sebai
- School of Medicine, California University of Science and Medicine, Colton, USA
| | - Lubhani Sharma
- Family Medicine, Dayanand Medical College and Hospital, Ludhiana, IND
| | - Mohamed Hassouba
- Pediatrics, SUNY Downstate Health Sciences University, Brooklyn, USA
| | - Ghazala S Virk
- Internal Medicine, Avalon University School of Medicine, Ohio, USA
| |
Collapse
|
206
|
Harte JV, Coleman-Vaughan C, Crowley MP, Mykytiv V. It's in the blood: a review of the hematological system in SARS-CoV-2-associated COVID-19. Crit Rev Clin Lab Sci 2023; 60:595-624. [PMID: 37439130 DOI: 10.1080/10408363.2023.2232010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to an unprecedented global healthcare crisis. While SARS-CoV-2-associated COVID-19 affects primarily the respiratory system, patients with COVID-19 frequently develop extrapulmonary manifestations. Notably, changes in the hematological system, including lymphocytopenia, neutrophilia and significant abnormalities of hemostatic markers, were observed early in the pandemic. Hematological manifestations have since been recognized as important parameters in the pathophysiology of SARS-CoV-2 and in the management of patients with COVID-19. In this narrative review, we summarize the state-of-the-art regarding the hematological and hemostatic abnormalities observed in patients with SARS-CoV-2-associated COVID-19, as well as the current understanding of the hematological system in the pathophysiology of acute and chronic SARS-CoV-2-associated COVID-19.
Collapse
Affiliation(s)
- James V Harte
- Department of Haematology, Cork University Hospital, Wilton, Cork, Ireland
- School of Biochemistry & Cell Biology, University College Cork, Cork, Ireland
| | | | - Maeve P Crowley
- Department of Haematology, Cork University Hospital, Wilton, Cork, Ireland
- Irish Network for Venous Thromboembolism Research (INViTE), Ireland
| | - Vitaliy Mykytiv
- Department of Haematology, Cork University Hospital, Wilton, Cork, Ireland
| |
Collapse
|
207
|
Samama M, Entezami F, Rosa DS, Sartor A, Piscopo RCCP, Andersen ML, Cunha-Filho JS, Jarmy-Di-Bella ZIK. COVID-19: A Challenge to the Safety of Assisted Reproduction. Sleep Med Clin 2023; 18:489-497. [PMID: 38501521 PMCID: PMC10288308 DOI: 10.1016/j.jsmc.2023.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
There is an increased risk of becoming pregnant through fertility treatments using assisted reproductive technology (ART) during the COVID-19 pandemic. The aim of this review is to gather comprehensive data from the existing literature on the potential risks of fertility management during the pandemic period, and outline strategies to mitigate them, with a focus on the hormonal and surgical procedures of ART. A comprehensive search of the scientific literature on COVID-19 in relation to fertility was conducted in the PubMed database using the keywords "coronavirus," "COVID-19," "SARS-CoV-2" and "pregnancy," "fertility," "urogenital system," "vertical transmission," "assisted human reproduction," "controlled ovarian stimulation," "oocyte retrieval," "in vitro fertilization," "hormones," "surgical procedures," "embryos," "oocytes," "sperm," "semen," "ovary," "testis," "ACE-2 receptor," "immunology," "cytokine storm," and "coagulation," from January 2020-July 2022. Published data on pregnancy and COVID-19, and the interaction of the urogenital system and SARS-CoV-2 is reported. The immunologic and prothrombotic profiles of patients with COVID-19, and their increased risks from controlled ovarian stimulation (COS) and ART surgeries, and how these procedures could facilitate COVID-19 and/or contribute to the severity of the disease by enhancing the cytokine storm are summarized. Strategies to prevent complications during COS that could increase the risks of the disease in pre-symptomatic patients are considered. The impact of SARS-CoV-2 on pre-symptomatic infertile patients presents a challenge to find ways to avoid the increased hormonal, immunologic, and prothrombotic risks presented by the use of COS in ART protocols during the COVID-19 outbreak. Safe ART procedures and recommendations are highlighted.
Collapse
Affiliation(s)
- Marise Samama
- Department of Gynecology, Federal University of São Paulo, São Paulo, Brazil; GERA Institute of Reproductive Medicine, São Paulo, Brazil.
| | - Frida Entezami
- American Hospital of Paris, IVF Unit, Neuilly-Sur-Seine, France
| | - Daniela S Rosa
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Amanda Sartor
- GERA Institute of Reproductive Medicine, São Paulo, Brazil; Department of Psychobiology, Federal University of São Paulo, São Paulo, Brazil
| | | | - Monica L Andersen
- Department of Psychobiology, Federal University of São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
208
|
Scheim DE, Vottero P, Santin AD, Hirsh AG. Sialylated Glycan Bindings from SARS-CoV-2 Spike Protein to Blood and Endothelial Cells Govern the Severe Morbidities of COVID-19. Int J Mol Sci 2023; 24:17039. [PMID: 38069362 PMCID: PMC10871123 DOI: 10.3390/ijms242317039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Consistent with well-established biochemical properties of coronaviruses, sialylated glycan attachments between SARS-CoV-2 spike protein (SP) and host cells are key to the virus's pathology. SARS-CoV-2 SP attaches to and aggregates red blood cells (RBCs), as shown in many pre-clinical and clinical studies, causing pulmonary and extrapulmonary microthrombi and hypoxia in severe COVID-19 patients. SARS-CoV-2 SP attachments to the heavily sialylated surfaces of platelets (which, like RBCs, have no ACE2) and endothelial cells (having minimal ACE2) compound this vascular damage. Notably, experimentally induced RBC aggregation in vivo causes the same key morbidities as for severe COVID-19, including microvascular occlusion, blood clots, hypoxia and myocarditis. Key risk factors for COVID-19 morbidity, including older age, diabetes and obesity, are all characterized by markedly increased propensity to RBC clumping. For mammalian species, the degree of clinical susceptibility to COVID-19 correlates to RBC aggregability with p = 0.033. Notably, of the five human betacoronaviruses, the two common cold strains express an enzyme that releases glycan attachments, while the deadly SARS, SARS-CoV-2 and MERS do not, although viral loads for COVID-19 and the two common cold infections are similar. These biochemical insights also explain the previously puzzling clinical efficacy of certain generics against COVID-19 and may support the development of future therapeutic strategies for COVID-19 and long COVID patients.
Collapse
Affiliation(s)
- David E Scheim
- US Public Health Service, Commissioned Corps, Inactive Reserve, Blacksburg, VA 24060, USA
| | - Paola Vottero
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, P.O. Box 208063, New Haven, CT 06520, USA
| | | |
Collapse
|
209
|
Hranjec T, Mayhew M, Rogers B, Solomon R, Hurst D, Estreicher M, Augusten A, Nunez A, Green M, Malhotra S, Katz R, Rosenthal A, Hennessy S, Pepe P, Sawyer R, Arenas J. Diagnosis and treatment of coagulopathy using thromboelastography with platelet mapping is associated with decreased risk of pulmonary failure in COVID-19 patients. Blood Coagul Fibrinolysis 2023; 34:508-516. [PMID: 37831624 DOI: 10.1097/mbc.0000000000001259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
INTRODUCTION Treatment of coronavirus disease 2019 (COVID-19) patients may require antithrombotic and/or anti-inflammatory medications. We hypothesized that individualized anticoagulant (AC) management, based on diagnosis of coagulopathy using thromboelastography with platelet mapping (TEG-PM), would decrease the frequency of pulmonary failure (PF) requiring mechanical ventilation (MV), mitigate thrombotic and hemorrhagic events, and, in-turn, reduce mortality. METHODS Hospital-admitted COVID-19 patients, age 18 or older, with escalating oxygen requirements were included. Prospective and supplemental retrospective chart reviews were conducted during a 2-month period. Patients were stratified into two groups based on clinician-administered AC treatment: TEG-PM guided vs. non-TEG guided. RESULTS Highly-elevated inflammatory markers (D-dimer, C-reactive protein, ferritin) were associated with poor prognosis but did not distinguish coagulopathic from noncoagulopathic patients. TEG-guided AC treatment was used in 145 patients vs. 227 treated without TEG-PM guidance. When managed by TEG-PM, patients had decreased frequency of PF requiring MV (45/145 [31%] vs. 152/227 [66.9%], P < 0.0001), fewer thrombotic events (2[1.4%] vs. 39[17.2%], P = 0.0019) and fewer hemorrhagic events (6[4.1%] vs. 24[10.7%], P = 0.0240), and had markedly reduced mortality (43[29.7%] vs. 142[62.6%], P < 0.0001). Platelet hyperactivity, indicating the need for antiplatelet medications, was identified in 75% of TEG-PM patients. When adjusted for confounders, empiric, indiscriminate AC treatment (not guided by TEG-PM) was shown to be an associated risk factor for PF requiring MV, while TEG-PM guided management was associated with a protective effect (odds ratio = 0.18, 95% confidence interval 0.08-0.4). CONCLUSIONS Following COVID-19 diagnosis, AC therapies based on diagnosis of coagulopathy using TEG-PM were associated with significantly less respiratory decompensation, fewer thrombotic and hemorrhagic complications, and improved likelihood of survival.
Collapse
Affiliation(s)
- Tjasa Hranjec
- Department of Surgery, Bronson Methodist Hospital
- Department of Surgery, Western Michigan University, Homer Stryker MD School of Medicine, Kalamazoo, Michigan
- Department of Surgery, Memorial Regional Hospital, Hollywood
| | - Mackenzie Mayhew
- Florida International University, Miami, Florida
- University of Virginia, Charlottesville, Virginia
| | | | - Rachele Solomon
- Department of Surgery, Memorial Regional Hospital, Hollywood
| | | | | | | | - Aaron Nunez
- Department of Medicine, Memorial Regional Hospital, Hollywood, Florida
| | - Melissa Green
- Department of Medicine, Memorial Regional Hospital, Hollywood, Florida
| | - Shivali Malhotra
- Department of Medicine, Memorial Regional Hospital, Hollywood, Florida
| | | | | | - Sara Hennessy
- Department of Surgery, University of Texas Southwestern Medical Center
| | - Paul Pepe
- Metropolitan Emergency Medical Services, Medical Directors Coalition Global Hdqtrs, Dallas, Texas, USA
| | - Robert Sawyer
- Department of Surgery, Bronson Methodist Hospital
- Department of Surgery, Western Michigan University, Homer Stryker MD School of Medicine, Kalamazoo, Michigan
| | - Juan Arenas
- Department of Surgery, Memorial Regional Hospital, Hollywood
| |
Collapse
|
210
|
Vanassche T, Engelen MM, Orlando C, Vandenbosch K, Gadisseur A, Hermans C, Jochmans K, Minon JM, Motte S, Peperstraete H, Péters P, Sprynger M, Lancellotti P, Dehaene I, Emonts P, Vandenbriele C, Verhamme P, Oury C. The 2023 Belgian clinical guidance on anticoagulation management in hospitalized and ambulatory COVID-19 patients. Acta Clin Belg 2023; 78:497-508. [PMID: 37548503 DOI: 10.1080/17843286.2023.2241692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023]
Abstract
COVID-19 is associated with an increased risk for thrombotic complications. The trials investigating the optimal thromboprophylactic dose are performed in challenging times and seemingly produce conflicting evidence. The burdensome circumstances, divergent endpoints, and different analytical approaches hamper comparison and extrapolation of available evidence. Most importantly, clinicians should provide thromboprophylaxis in hospitalized COVID-19 patients while (re)assessing bleeding and thrombotic risk frequently. The COVID-19 Thromboprophylaxis Working Group of the BSTH updated its guidance document. It aims to summarize the available evidence critically and to guide clinicians in providing the best possible thromboprophylaxis.
Collapse
Affiliation(s)
- Thomas Vanassche
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Matthias M Engelen
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Christelle Orlando
- Department of Haematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Kristel Vandenbosch
- Department of Laboratory Haematology, CHU University Hospital of Liege, Liege, Belgium
| | - Alain Gadisseur
- Department of Haematology, Antwerp University Hospital, Antwerp, Belgium
| | | | - Kristin Jochmans
- Department of Haematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Jean-Marc Minon
- Department of Laboratory Medicine, Thrombosis-Haemostasis and Transfusion Unit, CHR Citadelle, Liege, Belgium
| | - Serge Motte
- Department of Vascular Diseases, Erasme University Hospital, Brussels, Belgium
| | | | - Pierre Péters
- Department of Laboratory Haematology, CHU University Hospital of Liege, Liege, Belgium
| | - Muriel Sprynger
- Department of Cardiology, CHU University Hospital of Liege, Liege, Belgium
| | | | - Isabelle Dehaene
- Vlaamse Vereniging Voor Obstetrie En Gynaecologie, Universitair Ziekenhuis Gent, Ghent, Belgium
| | - Patrick Emonts
- Groupement des Gynecologues Obstetriciens de Langue Francaise de Belgique, CHU University Hospital of Liege, Liège, Belgium
| | | | - Peter Verhamme
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Cecile Oury
- Laboratory of Cardiology, GIGA Institute, University of Liege, Liege, Belgium
| |
Collapse
|
211
|
Guevara S, Miyara SJ, Aronsohn J, Homsi JT, McCann-Molmenti A, Mumford JM, Keber B, Shore-Lesserson L, Morales L, Metz CN, Cho YM, Molmenti CLS, Loto R, Pesce MM, Zafeiropoulos S, Giannis D, Pipolo DO, Jacque F, Montorfano L, Shinozaki K, Shoaib M, Choudhary RC, Nishikimi M, Takegawa R, Endo Y, Hayashida K, Fontan FM, Becker LB, Molmenti EP. COVID-19-Associated Portal Vein Thrombosis Post-Cholecystitis. Int J Angiol 2023; 32:262-268. [PMID: 37927847 PMCID: PMC10624542 DOI: 10.1055/s-0042-1743409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
This case study describes a 45-year-old Caucasian male with a past medical history of obesity, hypertension, and non-insulin-dependent diabetes mellitus, who in the setting of coronavirus disease 2019 (COVID-19) pneumonia, developed portal vein thrombosis (PVT) presenting as an acute abdomen after hospital discharge from a cholecystitis episode. PVT is a very infrequent thromboembolic condition, classically occurring in patients with systemic conditions such as cirrhosis, malignancy, pancreatitis, diverticulitis, autoimmunity, and thrombophilia. PVT can cause serious complications, such as intestinal infarction, or even death, if not promptly treated. Due to the limited number of reports in the literature describing PVT in the COVID-19 setting, its prevalence, natural history, mechanism, and precise clinical features remain unknown. Therefore, clinical suspicion should be high for PVT, in any COVID-19 patient who presents with abdominal pain or associated signs and symptoms. To the best of our knowledge, this is the first report of COVID-19-associated PVT causing extensive thrombosis in the portal vein and its right branch, occurring in the setting of early-stage cirrhosis after a preceding episode of cholecystitis.
Collapse
Affiliation(s)
- Sara Guevara
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Department of Family Medicine, Glen Cove Hospital, Glen Cove, New York
| | - Santiago J. Miyara
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York
| | - Judith Aronsohn
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Department of Anesthesiology, North Shore University Hospital, Manhasset, New York
| | - Joseph T. Homsi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - James M. Mumford
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Department of Family Medicine, Glen Cove Hospital, Glen Cove, New York
| | - Barbara Keber
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Department of Family Medicine, Glen Cove Hospital, Glen Cove, New York
| | - Linda Shore-Lesserson
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Department of Anesthesiology, North Shore University Hospital, Manhasset, New York
| | - Luis Morales
- Department of Surgery, North Shore University Hospital, Manhasset, New York
| | - Christine N. Metz
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York
| | - Young Min Cho
- Department of Internal Medicine, Northeast Georgia Medical Center, Gainesville, Georgia
| | | | - Rodrigo Loto
- Department of Radiology, Delta Sanatorium, Rosario, Santa Fe, ARG
| | - Martin M. Pesce
- Department of Radiology, Delta Sanatorium, Rosario, Santa Fe, ARG
| | - Stefanos Zafeiropoulos
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York
| | | | - Derek O. Pipolo
- Department of Surgery, North Shore University Hospital, Manhasset, New York
| | - Francky Jacque
- Department of Surgery, North Shore University Hospital, Manhasset, New York
| | | | - Koichiro Shinozaki
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Muhammad Shoaib
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Rishabh C. Choudhary
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Mitsuaki Nishikimi
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Ryosuke Takegawa
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Yusuke Endo
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Kei Hayashida
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Fermin M. Fontan
- Department of Surgery, Memorial Medical Center, Las Cruces, New Mexico
| | - Lance B. Becker
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York
| | - Ernesto P. Molmenti
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Surgery, North Shore University Hospital, Manhasset, New York
| |
Collapse
|
212
|
Bakr OB, El-Feky AH, Abdelazim IA, El-Skaan RG. Menstrual changes after the thrombo-prophylaxis or anticoagulants used during the COVID-19 infection. PRZEGLAD MENOPAUZALNY = MENOPAUSE REVIEW 2023; 22:179-185. [PMID: 38239401 PMCID: PMC10793612 DOI: 10.5114/pm.2023.133594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/10/2023] [Indexed: 01/22/2024]
Abstract
Introduction To detect the menstrual changes after the thrombo-prophylaxis or anticoagulants used during the COVID-19 infection. Material and methods A total of 176 diagnosed with COVID-19 infection, were included in this retrospective study after giving informed consent. Participants were asked to complete an online questionnaire, and the collected participants` data were analysed using the χ2 test to detect the menstrual changes after the thrombo-prophylaxis or anticoagulants used during the COVID-19 infection. Results The number of participants` number who reported menstrual flow for 2 to < 5 days, and menstrual flow > 7 days after the COVID-19 infection [31/176 (17.6%), and 42/176 (23.9%), respectively] was significantly higher compared to the number of participants` who reported menstrual flow for 2 to < 5 days, and menstrual flow > 7 days before the COVID-19 infection [12/176 (6.8%), and 15/176 (8.5%), respectively], (p = 0.005, and 0.0009, respectively). The use of thrombo-prophylaxis or anticoagulants during the COVID-19 infection was also associated with significant menstrual pattern changes (37.8% increased menstrual flow, 18.5% menstrual flow for 2 to < 5 days, 59.7% menstrual flow > 7 days, 5.9% contact bleeding, and 6.7% abnormal menstrual pattern for one cycle). Conclusions Significant menstrual changes were observed in this study after the COVID-19 infection infection (17.6% reported menstrual flow for 2 to < 5 days, and 23.9% reported menstrual flow > 7 days). The use of thrombo-prophylaxis or anticoagulants during the COVID-19 infection infection was associated with significant menstrual changes (37.8% increased menstrual flow, 18.5% menstrual flow for 2 to < 5 days, 59.7% menstrual flow > 7 days, 5.9% contact bleeding, and 6.7% abnormal menstrual pattern for one cycle).
Collapse
Affiliation(s)
- Omnia B. Bakr
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Alaa H. El-Feky
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ibrahim A. Abdelazim
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Rania G. El-Skaan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
213
|
Gangi-Burton A, Chan N, Ashok AH, Nair A. Simple demographic, laboratory and chest radiograph variables can identify COVID-19 patients with pulmonary thromboembolism: a retrospective multicentre United Kingdom study. Br J Radiol 2023; 96:20230082. [PMID: 37747264 PMCID: PMC10646650 DOI: 10.1259/bjr.20230082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/20/2023] [Accepted: 04/10/2023] [Indexed: 09/26/2023] Open
Abstract
OBJECTIVES To (1) identify discriminatory demographic, laboratory and initial CXR findings; (2) explore correlation between D-dimer and radiographic severity scores; and (3) assess accuracy of published D-dimer thresholds to identify pulmonary thromboembolism (PTE) in COVID-19 patients. METHODS Retrospective study including all COVID-19 patients admitted from 1st to 30th April 2020 meeting inclusion criteria from 25 (blinded) hospitals. Demographics, blood results, CXR and CTPA findings were compared between positive and negative PTE cohorts using uni- and multivariable logistic regression. Published D-dimer cut-offs were applied. RESULTS 389 patients were included [median age 63; 237 males], of which 26.2% had a PTE. Significant univariable discriminators for PTE were peak D-dimer, sex, neutrophil count at the time of the D-dimer and at admission, abnormal CXR, and CXR zonal severity score. Only neutrophil count at peak D-dimer remained significant for predicting PTE on multivariable analysis (p = 0.008). When compared with the published literature, sensitivity for PTE were lower than those published at all cut-off values, however specificity at different cut-offs was variable. CONCLUSIONS In this multicentre COVID-19 cohort, univariable admission factors that could indicate pulmonary thromboembolism were male sex, high neutrophil count and abnormal CXR with a greater CXR zonal severity score. The accuracy levels of published D-dimer thresholds were not reproducible in our population. ADVANCES IN KNOWLEDGE This is a large multicentre study looking at the discriminatory value of simple variables to determine if a patient with COVID-19 has PTE or not, in addition to comparing D-dimer cut off values against published values.
Collapse
Affiliation(s)
- Anmol Gangi-Burton
- Department of Radiology, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Nathan Chan
- Department of Interventional Neuroradiology, The Royal London Hospital, London, United Kingdom
| | - Abhishekh H Ashok
- Department of Radiology, Addenbrooke’s Hospital, Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
| | - Arjun Nair
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
214
|
Wolny M, Dittrich M, Knabbe C, Birschmann I. Immature platelets in COVID-19. Platelets 2023; 34:2184183. [PMID: 36883692 DOI: 10.1080/09537104.2023.2184183] [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: 03/09/2023]
Abstract
Platelets play a critical role in immune response. Coronavirus disease 2019 (COVID-19) patients with a severe course often show pathological coagulation parameters including thrombocytopenia, and at the same time the proportion of immature platelets increases. In this study, the platelet count and the immature platelet fraction (IPF) of hospitalized patients with different oxygenation requirements was investigated daily over a course of 40 days. In addition, the platelet function of COVID-19 patients was analyzed. It was found that the number of platelets in patients with the most severe course (intubation and extracorporeal membrane oxygenation (ECMO)) was significantly lower (111.5 ∙ 106 /mL) than in the other groups (mild (no intubation, no ECMO): 203.5 ∙ 106 /mL, p < .0001, moderate (intubation, no ECMO): 208.0 ∙ 106 /mL, p < .0001). IPF tended to be elevated (10.9%). Platelet function was reduced. Differentiation by outcome revealed that the deceased patients had a highly significant lower platelet count and higher IPF (97.3 ∙ 106 /mL, p < .0001, 12.2%, p = .0003).
Collapse
Affiliation(s)
- Monika Wolny
- Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum NRW, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Deutschland
| | - Marcus Dittrich
- Institut für Humangenetik und Bioinformatik, Universität Würzburg, Würzburg, Deutschland
| | - Cornelius Knabbe
- Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum NRW, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Deutschland
| | - Ingvild Birschmann
- Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum NRW, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Deutschland
| |
Collapse
|
215
|
Landi A, Morici N, Vranckx P, Frigoli E, Bonacchini L, Omazzi B, Tresoldi M, Camponovo C, Moccetti T, Valgimigli M. Edoxaban and/or colchicine in outpatients with COVID-19: rationale and design of the CONVINCE trial. J Cardiovasc Med (Hagerstown) 2023; 24:920-930. [PMID: 37942793 DOI: 10.2459/jcm.0000000000001556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
BACKGROUND An excessive inflammatory response and a hypercoagulable state are not infrequent in patients with coronavirus disease-2019 (COVID-19) and are associated with adverse clinical outcomes. However, the optimal treatment strategy for COVID-19 patients managed in the out-of-hospital setting is still uncertain. DESIGN The CONVINCE (NCT04516941) is an investigator-initiated, open-label, blinded-endpoint, 2 × 2 factorial design randomized trial aimed at assessing two independently tested hypotheses (anticoagulation and anti-inflammatory ones) in COVID-19 patients. Adult symptomatic patients (≥18 years of age) within 7 days from reverse transcription-PCR (RT-PCR) diagnosis of SARS-CoV-2 infection managed at home or in nursery settings were considered for eligibility. Eligible patients fulfilling all inclusion and no exclusion criteria were randomized to edoxaban versus no treatment (anticoagulation hypothesis) and colchicine versus no treatment (anti-inflammatory hypothesis) in a 1 : 1:1 : 1 ratio. The study had two co-primary endpoints (one for each randomization), including the composite of major vascular thrombotic events at 25 ± 3 days for the anticoagulation hypothesis and the composite of SARS-CoV-2 detection rates at 14 ± 3 days by RT-PCR or freedom from death or hospitalizations (anti-inflammatory hypothesis). Study endpoints will be adjudicated by a blinded Clinical Events Committee. With a final sample size of 420 patients, this study projects an 80% power for each of the two primary endpoints appraised separately. CONCLUSION The CONVINCE trial aims at determining whether targeting anticoagulation and/or anti-inflammatory pathways may confer benefit in COVID-19 patients managed in the out-of-hospital setting. TRIAL REGISTRATION ClinicalTrials.gov number, NCT04516941.
Collapse
Affiliation(s)
- Antonio Landi
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale (EOC)
- Department of Biomedical Sciences, University of Italian Switzerland, Lugano, Switzerland
| | - Nuccia Morici
- IRCCS S. Maria Nascente - Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Pascal Vranckx
- the Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Enrico Frigoli
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale (EOC)
| | - Luca Bonacchini
- Emergency Department, ASST Great Metropolitan Hospital Niguarda, Milan
| | - Barbara Omazzi
- Emergency Unit, ASST Rhodense, Garbagnate Milanese, Italy
| | - Moreno Tresoldi
- Unit of General Medicine and Advanced Care, IRCCS San Raffaele Hospital, Milan
| | - Claudio Camponovo
- Department of Anesthesiology, Clinica Ars Medica, Genolier Swiss Medical Network, Gravesano
| | | | - Marco Valgimigli
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale (EOC)
- Department of Biomedical Sciences, University of Italian Switzerland, Lugano, Switzerland
- University of Bern, Bern, Switzerland
| |
Collapse
|
216
|
Lillicrap D, Morrissey JH. Looking Back-And Forward. J Thromb Haemost 2023; 21:3285-3286. [PMID: 38000849 DOI: 10.1016/j.jtha.2023.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2023]
Affiliation(s)
- David Lillicrap
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - James H Morrissey
- Departments of Biological Chemistry & Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
217
|
Zelaya H, Arellano-Arriagada L, Fukuyama K, Matsumoto K, Marranzino G, Namai F, Salva S, Alvarez S, Agüero G, Kitazawa H, Villena J. Lacticaseibacillus rhamnosus CRL1505 Peptidoglycan Modulates the Inflammation-Coagulation Response Triggered by Poly(I:C) in the Respiratory Tract. Int J Mol Sci 2023; 24:16907. [PMID: 38069229 PMCID: PMC10707514 DOI: 10.3390/ijms242316907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Lacticaseibacillus rhamnosus CRL1505 beneficially modulates the inflammation-coagulation response during respiratory viral infections. This study evaluated the capacity of the peptidoglycan obtained from the CRL1505 strain (PG-Lr1505) to modulate the immuno-coagulative response triggered by the viral pathogen-associated molecular pattern poly(I:C) in the respiratory tract. Adult BALB/c mice were nasally treated with PG-Lr1505 for two days. Treated and untreated control mice were then nasally challenged with poly(I:C). Mice received three doses of poly(I:C) with a 24 h rest period between each administration. The immuno-coagulative response was studied after the last administration of poly(I:C). The challenge with poly(I:C) significantly increased blood and respiratory pro-inflammatory mediators, decreased prothrombin activity (PT), and increased von Willebrand factor (vWF) levels in plasma. Furthermore, tissue factor (TF), tissue factor pathway inhibitor (TFPI), and thrombomodulin (TM) expressions were increased in the lungs. PG-Lr1505-treated mice showed significant modulation of hemostatic parameters in plasma (PT in %, Control = 71.3 ± 3.8, PG-Lr1505 = 94.0 ± 4.0, p < 0.01) and lungs. Moreover, PG-Lr1505-treated mice demonstrated reduced TF in F4/80 cells from lungs, higher pro-inflammatory mediators, and increased IL-10 compared to poly(I:C) control mice (IL-10 in pg/mL, Control = 379.1 ± 12.1, PG-Lr1505 = 483.9 ± 11.3, p < 0.0001). These changes induced by PG-Lr1505 correlated with a significant reduction in lung tissue damage. Complementary in vitro studies using Raw 264.7 cells confirmed the beneficial effect of PG-Lr1505 on poly(I:C)-induced inflammation, since increased IL-10 expression, as well as reduced damage, production of inflammatory mediators, and hemostatic parameter expressions were observed. In addition, protease-activated receptor-1 (PAR1) activation in lungs and Raw 264.7 cells was observed after TLR3 stimulation, which was differentially modulated by PG-Lr1505. The peptidoglycan from L. rhamnosus CRL1505 is able to regulate inflammation, the procoagulant state, and PAR1 activation in mice and macrophages in the context of the activation of TLR3 signaling pathways, contributing to a beneficial modulation of inflammation-hemostasis crosstalk.
Collapse
Affiliation(s)
- Hortensia Zelaya
- Institute of Applied Biochemistry, Tucuman University, Tucuman 4000, Argentina; (H.Z.); (S.A.); (G.A.)
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (L.A.-A.); (G.M.); (S.S.)
| | - Luciano Arellano-Arriagada
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (L.A.-A.); (G.M.); (S.S.)
| | - Kohtaro Fukuyama
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan; (K.F.); (K.M.); (F.N.)
| | - Kaho Matsumoto
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan; (K.F.); (K.M.); (F.N.)
| | - Gabriela Marranzino
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (L.A.-A.); (G.M.); (S.S.)
- Facultad de Ciencias de la Salud, Universidad del Norte Santo Tomás de Aquino (UNSTA), Tucuman 4000, Argentina
| | - Fu Namai
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan; (K.F.); (K.M.); (F.N.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan
| | - Susana Salva
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (L.A.-A.); (G.M.); (S.S.)
| | - Susana Alvarez
- Institute of Applied Biochemistry, Tucuman University, Tucuman 4000, Argentina; (H.Z.); (S.A.); (G.A.)
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (L.A.-A.); (G.M.); (S.S.)
| | - Graciela Agüero
- Institute of Applied Biochemistry, Tucuman University, Tucuman 4000, Argentina; (H.Z.); (S.A.); (G.A.)
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan; (K.F.); (K.M.); (F.N.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (L.A.-A.); (G.M.); (S.S.)
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan; (K.F.); (K.M.); (F.N.)
| |
Collapse
|
218
|
Kafan S, Fattahi MR, Akhbari Shojaei M, Hossein Nezhad A, Imankhan M, Jahansouz D, Montazeri M, Hadadi A, Fattahi S, Iranmehr A, Pazoki M, Rahimzadeh H. Comparing Therapeutic versus Prophylactic Enoxaparin Therapy in Severe COVID-19 Patients: A Randomized Clinical Trial. Med J Islam Repub Iran 2023; 37:129. [PMID: 38318404 PMCID: PMC10843207 DOI: 10.47176/mjiri.37.129] [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: 12/14/2022] [Indexed: 02/07/2024] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) has been associated with a hypercoagulopathy state; however, the efficacy of different anticoagulant regimens in preventing thrombotic events is not clear. We aimed to compare therapeutic versus prophylactic enoxaparin therapy in severe COVID-19 patients. Methods In this single-center, open-label, randomized controlled trial, adult patients with severe COVID-19 presentations and an increased D-dimer level of more than 4 times the normal upper limit were randomly assigned to receive either prophylactic or therapeutic dose of enoxaparin. All patients were observed for at least 4 months regarding the overall survival as the primary outcome. Hospitalization duration, the need for intensive care unit (ICU) admission, the need for mechanical ventilation, and major adverse events (MAEs) were also analyzed as the secondary outcomes. Survival analysis was done via Kaplan-Meier curves and the Log-rank test. Cox regression was used, adjusting for baseline variables. Results Overall, 237 patients (152 men and 85 women) were randomized to either arm (121 to prophylactic and 116 to therapeutic groups). The mortality rate was 27 (22.3%) and 52 (44.8%) in prophylactic and therapeutic arms, respectively. Prophylactic enoxaparin was associated with better survival in the log-rank test (P < 0.001; HR, 0.42). Additionally, a significantly lower rate of ICU admission, a lower rate of MAEs, and shorter hospitalization were observed in the prophylactic arm (P < 0.001, P = 0.009, and P = 0.028, respectively). Conclusion The results of the current study were in favor of anticoagulant treatment with prophylactic doses of enoxaparin. Still, due to the limitations of this paper, we suggest that these findings be treated cautiously.
Collapse
Affiliation(s)
- Samira Kafan
- Department of Internal Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Fattahi
- Department of Internal Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Akhbari Shojaei
- Department of Internal Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Aida Hossein Nezhad
- School of Medicine, Islamic Azad University, Tonekabon Medical Branch, Tonekabon, Iran
| | - Mahshid Imankhan
- School of Medicine, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Davoud Jahansouz
- Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mahnaz Montazeri
- Department of Infectious Disease, Tehran University of Medical Sciences, Tehran, Iran
| | - Azar Hadadi
- Department of Internal Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Infectious Disease, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Fattahi
- Department of Clinical Psychology, Tehran University of Medical Sciences, Tehran, Iran
| | - Arad Iranmehr
- Neurosurgery Department, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Pazoki
- Department of Internal Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Hormat Rahimzadeh
- Department of Nephrology Disease, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
219
|
Liu XQ, Lu GZ, Yin DL, Kang YY, Zhou YY, Wang YH, Xu J. Analysis of clinical characteristics and risk factors between elderly patients with severe and nonsevere Omicron variant infection. World J Clin Infect Dis 2023; 13:37-48. [DOI: 10.5495/wjcid.v13.i4.37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/02/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to millions of confirmed cases and deaths worldwide. Elderly patients are at high risk of developing and dying from COVID-19 due to advanced age, decreased immune function, intense inflammatory response, and comorbidities. Shanghai has experienced a wave of infection with Omicron, a new variant of SARS-CoV-2, since March 2022. There is a pressing need to identify clinical features and risk factors for disease progression among elderly patients with Omicron infection to provide solid evidence for clinical policy-makers, public health officials, researchers, and the general public.
AIM To investigate clinical characteristic differences and risk factors between elderly patients with severe and nonsevere Omicron SARS-CoV-2 variant infection.
METHODS A total of 328 elderly patients with COVID-19 admitted to the Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine from April 2022 to June 2022 were enrolled and divided into a severe group (82 patients) and a nonsevere group (246 patients) according to the diagnosis and treatment protocol of COVID-19 (version 7). The clinical data and laboratory results of both groups were collected and compared. A chi-square test, t test, Mann-Whitney U test, hierarchical log-rank test, univariate and multivariate logistic regression, and hierarchical analyses were used to determine significant differences.
RESULTS The severe group was older (84 vs 74 years, P < 0.001), included more males (57.3% vs 43.9%, P = 0.037), had a lower vaccination rate (P < 0.001), and had a higher proportion of comorbidities, including chronic respiratory disease (P = 0.001), cerebral infarction (P < 0.001), chronic kidney disease (P = 0.002), and neurodegenerative disease (P < 0.001), than the nonsevere group. In addition, severe disease patients had a higher inflammatory index (P < 0.001), greater need for symptomatic treatment (P < 0.001), longer hospital stay (P = 0.011), extended viral shedding time (P = 0.014), and higher mortality than nonsevere disease patients (P < 0.001). No difference was observed in the application of Paxlovid in the severe and nonsevere groups (P = 0.817). Oxygen saturation, cerebral infarction, and D-dimer were predictive factors for developing severe disease in patients with COVID-19, with D-dimer having an excellent role (area under the curve: 90.1%, 95%CI: 86.1-94.0%). In addition, D-dimer was a risk factor for developing severe COVID-19 according to multivariate stratified analysis.
CONCLUSION The clinical course of severe COVID-19 is complex, with a higher need for symptomatic treatment. D-dimer is a suitable biomarker for identifying patients at risk for developing severe COVID-19.
Collapse
Affiliation(s)
- Xiao-Qin Liu
- Department of Infectious Disease, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Guan-Zhu Lu
- Department of Infectious Disease, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Dong-Lin Yin
- Department of Infectious Disease, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yao-Yue Kang
- Department of Infectious Disease, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yuan-Yuan Zhou
- Department of Infectious Disease, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yu-Huan Wang
- Department of Infectious Disease, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jie Xu
- Department of Infectious Disease, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
220
|
Zhang Z, Zhang N, Lu X, Zhou M, Yan X, Gu W, Yang J, Zhang Q, Zhang C, Gong Y, Jia M, Zhang X, Ning P, Liu M, Li X, Shi X, Liu W, Gao GF, Ning G, Wang J, Bi Y. Anti-infection effects of heparin on SARS-CoV-2 in a diabetic mouse model. Zool Res 2023; 44:1003-1014. [PMID: 37759335 PMCID: PMC10802103 DOI: 10.24272/j.issn.2095-8137.2023.108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can result in more severe syndromes and poorer outcomes in patients with diabetes and obesity. However, the precise mechanisms responsible for the combined impact of corona virus disease 2019 (COVID-19) and diabetes have not yet been elucidated, and effective treatment options for SARS-CoV-2-infected diabetic patients remain limited. To investigate the disease pathogenesis, K18-hACE2 transgenic (hACE2 Tg) mice with a leptin receptor deficiency (hACE2-Lepr -/-) or high-fat diet (hACE2-HFD) background were generated. The two mouse models were intranasally infected with a 5×10 5 median tissue culture infectious dose (TCID 50) of SARS-CoV-2, with serum and lung tissue samples collected at 3 days post-infection. The hACE2-Lepr -/- mice were then administered a combination of low-molecular-weight heparin (LMWH) (1 mg/kg or 5 mg/kg) and insulin via subcutaneous injection prior to intranasal infection with 1×10 4 TCID 50 of SARS-CoV-2. Daily drug administration continued until the euthanasia of the mice. Analyses of viral RNA loads, histopathological changes in lung tissue, and inflammation factors were conducted. Results demonstrated similar SARS-CoV-2 susceptibility in hACE2 Tg mice under both lean (chow diet) and obese (HFD) conditions. However, compared to the hACE2-Lepr +/+ mice, hACE2-Lepr -/- mice exhibited more severe lung injury, enhanced expression of inflammatory cytokines and hypoxia-inducible factor-1α, and increased apoptosis. Moreover, combined LMWH and insulin treatment effectively reduced disease progression and severity, attenuated lung pathological changes, and mitigated inflammatory responses. In conclusion, pre-existing diabetes can lead to more severe lung damage upon SARS-CoV-2 infection, and LMWH may be a valuable therapeutic approach for managing COVID-19 patients with diabetes.
Collapse
Affiliation(s)
- Zhongyun Zhang
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Ning Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Xuancheng Lu
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Min Zhou
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoxiang Yan
- Department of Cardiology, Institute of Cardiovascular Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weiqiong Gu
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Jingru Yang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qin Zhang
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Cheng Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Yuhuan Gong
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Mingjun Jia
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoyu Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Peng Ning
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Mei Liu
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Xiaoyan Li
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Xiaomeng Shi
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - George F Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guang Ning
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China. E-mail:
| | - Yuhai Bi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- University of Chinese Academy of Sciences, Beijing 100049, China. E-mail:
| |
Collapse
|
221
|
Alizad G, Ayatollahi AA, Shariati Samani A, Samadizadeh S, Aghcheli B, Rajabi A, Nakstad B, Tahamtan A. Hematological and Biochemical Laboratory Parameters in COVID-19 Patients: A Retrospective Modeling Study of Severity and Mortality Predictors. BIOMED RESEARCH INTERNATIONAL 2023; 2023:7753631. [PMID: 38027038 PMCID: PMC10676280 DOI: 10.1155/2023/7753631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/08/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023]
Abstract
Background It is well known that laboratory markers could help in identifying risk factors of severe illness and predicting outcomes of diseases. Here, we performed a retrospective modeling study of severity and mortality predictors of hematological and biochemical laboratory parameters in Iranian COVID-19 patients. Methods Data were obtained retrospectively from medical records of 564 confirmed Iranian COVID-19 cases. According to the disease severity, the patients were categorized into two groups (severe or nonsevere), and based on the outcome of the disease, patients were divided into two groups (recovered or deceased). Demographic and laboratory data were compared between groups, and statistical analyses were performed to define predictors of disease severity and mortality in the patients. Results The study identified a panel of hematological and biochemical markers associated with the severe outcome of COVID-19 and constructed different predictive models for severity and mortality. The disease severity and mortality rate were significantly higher in elderly inpatients, whereas gender was not a determining factor of the clinical outcome. Age-adjusted white blood cells (WBC), platelet cells (PLT), neutrophil-to-lymphocyte ratio (NLR), red blood cells (RBC), hemoglobin (HGB), hematocrit (HCT), erythrocyte sedimentation rate (ESR), mean corpuscular hemoglobin (MCHC), blood urea nitrogen (BUN), and creatinine (Cr) also showed high accuracy in predicting severe cases at the time of hospitalization, and logistic regression analysis suggested grouped hematological parameters (age, WBC, NLR, PLT, HGB, and international normalized ratio (INR)) and biochemical markers (age, BUN, and lactate dehydrogenase (LDH)) as the best models of combined laboratory predictors for severity and mortality. Conclusion The findings suggest that a panel of several routine laboratory parameters recorded on admission could be helpful for clinicians to predict and evaluate the risk of disease severity and mortality in COVID-19 patients.
Collapse
Affiliation(s)
- Ghazaleh Alizad
- Department of Immunology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Asghar Ayatollahi
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Saeed Samadizadeh
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Bahman Aghcheli
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abdolhalim Rajabi
- Environmental Health Research Center, Biostatistics & Epidemiology Department, Faculty of Health, Golestan University of Medical Sciences, Gorgan, Iran
| | - Britt Nakstad
- Division of Paediatric and Adolescent Medicine, University of Oslo, Oslo, Norway
- Department of Paediatrics and Adolescent Health, University of Botswana, Gaborone, Botswana
| | - Alireza Tahamtan
- School of International, Golestan University of Medical Sciences, Gorgan, Iran
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
222
|
Carestia A, Godin LC, Jenne CN. Step up to the platelet: Role of platelets in inflammation and infection. Thromb Res 2023; 231:182-194. [PMID: 36307228 DOI: 10.1016/j.thromres.2022.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/18/2022]
Abstract
Platelets are anucleated cells derived from megakaryocytes that are primarily responsible for hemostasis. However, in recent years, these cytoplasts have become increasingly recognized as immune cells, able to detect, interact with, and kill pathogens. As platelets are involved in both immunity and coagulation, they have a central role in immunothrombosis, a physiological process in which immune cells induce the formation of microthrombi to both prevent the spread of pathogens, and to help facilitate clearance. In this review, we will highlight the role of platelets as key players in the inflammatory and innate immune response against bacterial and viral infection, including direct and indirect interactions with pathogens and other immune cells.
Collapse
Affiliation(s)
- Agostina Carestia
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada.
| | - Laura C Godin
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada.
| | - Craig N Jenne
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada.
| |
Collapse
|
223
|
Lima WDS, Soares MHP, Paschoal EHA, Paschoal JKSF, Paschoal FM, Bor-Seng-Shu E. Intracranial hemorrhages in patients with COVID-19: a systematic review of the literature, regarding six cases in an Amazonian population. ARQUIVOS DE NEURO-PSIQUIATRIA 2023; 81:989-999. [PMID: 38035584 PMCID: PMC10689113 DOI: 10.1055/s-0043-1772834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 05/01/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has emerged as a public health emergency worldwide, predominantly affecting the respiratory tract. However, evidence supports the involvement of extrapulmonary sites, including reports of intracranial hemorrhages. OBJECTIVE To describe six original cases and review the literature on intracranial hemorrhages in patients diagnosed with COVID-19 by molecular methods. METHODS A systematic literature review was performed on MEDLINE, PubMed, and NCBI electronic databases to identify eligible studies. Of the total 1,624 articles retrieved, only 53 articles met the inclusion criteria. RESULTS The overall incidence of intracranial hemorrhage in patients hospitalized for COVID-19 was 0.26%. In this patient group, the mean age was 60 years, and the majority were male (68%) with initial respiratory symptoms (73%) and some comorbidity. Before the diagnosis of hemorrhage, 43% of patients were using anticoagulants, 47.3% at therapeutic doses. The intraparenchymal (50%) was the most affected compartment, followed by the subarachnoid (34%), intraventricular (11%), and subdural (7%). There was a predominance of lobar over non-lobar topographies. Multifocal or multicompartmental hemorrhages were described in 25% of cases. Overall mortality in the cohort studies was 44%, while around 55% of patients were discharged from hospital. CONCLUSION Despite the unusual association, the combination of these two diseases is associated with high rates of mortality and morbidity, as well as more severe clinicoradiological presentations. Further studies are needed to provide robust evidence on the exact pathophysiology behind the occurrence of intracranial hemorrhages after COVID-19 infection.
Collapse
Affiliation(s)
- William de Sousa Lima
- Universidade Federal do Pará, Faculdade de Medicina, Departamento de Neurologia do Hospital Universitário João de Barros Barreto, Belém PA, Brazil.
| | - Marcelo Henrique Pereira Soares
- Universidade Federal do Pará, Faculdade de Medicina, Departamento de Neurologia do Hospital Universitário João de Barros Barreto, Belém PA, Brazil.
| | - Eric Homero Albuquerque Paschoal
- Universidade Federal do Pará, Faculdade de Medicina, Departamento de Neurologia do Hospital Universitário João de Barros Barreto, Belém PA, Brazil.
| | | | - Fernando Mendes Paschoal
- Universidade Federal do Pará, Faculdade de Medicina, Departamento de Neurologia do Hospital Universitário João de Barros Barreto, Belém PA, Brazil.
| | - Edson Bor-Seng-Shu
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia do Hospital das Clínicas, São Paulo SP, Brazil.
| |
Collapse
|
224
|
Scalambrino E, Clerici M, Scardo S, Capecchi M, Della Noce C, Testa S, Peyvandi F, Tripodi A. COVID-19. Comparison of D-dimer levels measured with 3 commercial platforms. Res Pract Thromb Haemost 2023; 7:102247. [PMID: 38193049 PMCID: PMC10772868 DOI: 10.1016/j.rpth.2023.102247] [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: 02/22/2023] [Revised: 09/13/2023] [Accepted: 10/19/2023] [Indexed: 01/10/2024] Open
Affiliation(s)
- Erica Scalambrino
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milano, Italy
| | - Marigrazia Clerici
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milano, Italy
| | - Sara Scardo
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milano, Italy
| | - Marco Capecchi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milano, Italy
| | | | | | - Flora Peyvandi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milano, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Armando Tripodi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milano, Italy
- Fondazione Luigi Villa, Milano, Italy
| |
Collapse
|
225
|
Sevilya Z, Kuzmina A, Cipok M, Hershkovitz V, Keidar-Friedman D, Taube R, Lev EI. Differential platelet activation through an interaction with spike proteins of different SARS-CoV-2 variants. J Thromb Thrombolysis 2023; 56:538-547. [PMID: 37736784 DOI: 10.1007/s11239-023-02891-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 09/23/2023]
Abstract
COVID-19 disease is associated with an increased risk of thrombotic complications, which contribute to high short-term mortality. Patients with COVID-19 demonstrate enhanced platelet turnover and reactivity, which may have a role in the development of thrombotic events and disease severity. Evidence has suggested direct interaction between SARS-CoV-2 and platelets, resulting in platelets activation. Here, we compare the effect of various SARS-CoV-2 spike variants on platelet activation. Engineered lentiviral particles were pseudotyped with spike SARS-CoV-2 variants and incubated with Platelet Rich Plasma obtained from healthy individuals. The pseudotyped SARS-CoV-2 exhibiting the wild-type Wuhan-Hu spike protein stimulated platelets to increase expression of the surface CD62P and activated αIIbβ3 markers by 3.5 ± 1.2 and 3.3 ± 0.7 fold, respectively (P = 0.004 and 0.003). The Delta variant induced much higher levels of platelet activation; CD62P expression was increased by 6.6 ± 2.2 fold and activated αIIbβ3 expression was increased by 5.0 ± 1.5 fold (P = 0.005 and 0.026, respectively). The Omicron BA.1 and the Alpha variants induced the lowest level of activation; CD62P expression was increased by 1.7 ± 0.4 and 1.6 ± 0.9 fold, respectively (P = 0.003 and 0.008), and activated αIIbβ3 expression by 1.8 ± 1.1 and 1.6 ± 0.8, respectively (P = 0.003 and 0.001). The Omicron BA.2 variant induced an increase of platelets activation comparable to the Wuhan-Hu (2.8 ± 1.2 and 2.1 ± 1.3 fold for CD62P and activated αIIbβ3 markers, respectively). The results obtained for various COVID-19 variants are in correlation with the clinical severity and mortality reported for these variants.
Collapse
Affiliation(s)
- Ziv Sevilya
- Cardiology Department, Assuta Ashdod Medical Center, Ashdod, Israel.
| | - Alona Kuzmina
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Michal Cipok
- Hematology Laboratory, Assuta Ashdod Medical Center, Ashdod, Israel
| | - Vera Hershkovitz
- Hematology Laboratory, Assuta Ashdod Medical Center, Ashdod, Israel
| | | | - Ran Taube
- The Shraga Segal Department of Microbiology Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Eli I Lev
- Cardiology Department, Assuta Ashdod Medical Center, Ashdod, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
226
|
Onishi T, Harada S, Shimo H, Tashiro Y, Soeda T, Nogami K. The in vitro effect of anticoagulant agents on coagulation and fibrinolysis in the presence of emicizumab in the plasmas from patients with haemophilia A. Haemophilia 2023; 29:1529-1538. [PMID: 37766492 DOI: 10.1111/hae.14877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
INTRODUCTION Emicizumab is used as hemostatic prophylaxis for patients with hemophilia A (PwHA), irrespective of the presence of inhibitors. Although bacterial infection can lead to a procoagulant state, there is limited information on coagulation and fibrinolysis potentials in emicizumab-treated PwHA and on the use of anticoagulants in such cases. AIM We examined whether anticoagulants affect the coagulation and fibrinolysis potentials in plasma from PwHA spiked with emicizumab. METHODS Plasma from PwHA was in vitro supplemented with emicizumab (50 μg/mL; emi-plasma) and anticoagulants (recombinant thrombomodulin (rTM), nafamostat mesylate (NM), unfractionated heparin (UFH), or low-molecular-weight heparin (LMH)). PwHA plasma spiked with rFVIII (1 IU/mL) was used as a reference (ref-plasma). The coagulation and fibrinolysis potentials in plasma was measured by thrombin and plasmin generation assay (T/P-GA) and clot-fibrinolysis waveform analysis (CFWA). RESULTS In T/P-GA and CFWA, coagulation potentials (maximum coagulation velocity; |min1|, and peak thrombin; Th-Peak) in plasma rose with increasing concentrations of emicizumab and rFVIII, but fibrinolytic potentials (peak plasmin; Plm-Peak, and maximum fibrinolytic velocity; |FL-min1|) remained unchanged. Adding rTM, NM, and UFH to emi-plasma suppressed coagulation and fibrinolysis potentials, similar to ref-plasma. Regarding the heparin, UFH and LMH inhibited the improved coagulation in emi-plasma. UFH inhibited fibrinolysis as well, but LMH did not. CONCLUSIONS Anticoagulants could exhibit the inhibitory effects on the coagulation and fibrinolysis potentials in plasma from PwHA spiked with emicizumab, similar to those in normal plasma.
Collapse
Affiliation(s)
- Tomoko Onishi
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - Suguru Harada
- Chugai Pharmaceutical Co., Ltd., Yokohama, Kanagawa, Japan
| | - Hanako Shimo
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | | | | | - Keiji Nogami
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
227
|
Quarterman C, Cole O. Anti-Xa assay monitoring for thromboprophylaxis in critical care patients with Covid-19 pneumonia. J Intensive Care Soc 2023; 24:37-38. [PMID: 37928075 PMCID: PMC10621526 DOI: 10.1177/1751143720966279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023] Open
Affiliation(s)
| | - Oana Cole
- Liverpool Heart and Chest NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
228
|
Tashkandi WA. Incidence and Risk Factors Associated with Thromboembolic Events among Patients with COVID-19 Inpatients: A Retrospective Study. Indian J Crit Care Med 2023; 27:830-836. [PMID: 37936799 PMCID: PMC10626239 DOI: 10.5005/jp-journals-10071-24575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/29/2023] [Indexed: 11/09/2023] Open
Abstract
Aims and objectives Despite thromboprophylaxis, some severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) patients develop thrombotic complications with poor prognosis. Our goal is to comprehensively assess the incidence, risk factors, and clinical outcomes associated with thromboembolic events (TE) among adult patients presenting with coronavirus disease-2019 (COVID-19). Materials and methods The study was conducted as an observational and retrospective study across COVID-19 patients (n = 207) in a tertiary care hospital in the Middle East and North Africa (MENA) region. Electronic health records were collected from the COVID-19 Database from April 2020 to December 2020 which included clinical history and TE. Results Fifty-six (27.05%) out of 207 patients (age: 54.42 ± 15.01 years) developed TE despite the anticoagulant therapy. The incidence of venous thromboembolism (VTE) was significantly higher for patients aged >50 years compared to <50 years (73.21% vs 26.79%, p < 0.05). There were no differences in the incidence of VTE between genders (p = 0.561). 165 patients (79.71%) received anticoagulant therapy, yet 48 (29%) developed TE. The most commonly used anticoagulant was low-molecular-weight heparin (LMWH, 47.34%). In spite of efficient treatment and medical management, the majority of patients with TE (45 out of 56 patients, 80.35%) experienced mortality. The comorbidities that significantly increase the risk of TE include hypertension (HTN) and ischemic heart disease (IHD). The laboratory parameters that were associated with an increased risk of VTE include ferritin, lactate dehydrogenase (LDH), and creatinine. Conclusion The COVID-19 patients develop thrombotic complications. Future studies should clarify the underlying mechanisms of TE and optimize the antithrombotic regimens in COVID-19 patients. How to cite this article Tashkandi WA. Incidence and Risk Factors Associated with Thromboembolic Events among Patients with COVID-19 Inpatients: A Retrospective Study. Indian J Crit Care Med 2023;27(11):830-836.
Collapse
Affiliation(s)
- Wail Abdulhafez Tashkandi
- Department of Surgery, Faculty of Medicine, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
229
|
Ortega-Paz L, Talasaz AH, Sadeghipour P, Potpara TS, Aronow HD, Jara-Palomares L, Sholzberg M, Angiolillo DJ, Lip GYH, Bikdeli B. COVID-19-Associated Pulmonary Embolism: Review of the Pathophysiology, Epidemiology, Prevention, Diagnosis, and Treatment. Semin Thromb Hemost 2023; 49:816-832. [PMID: 36223804 DOI: 10.1055/s-0042-1757634] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
COVID-19 is associated with endothelial activation in the setting of a potent inflammatory reaction and a hypercoagulable state. The end result of this thromboinflammatory state is an excess in thrombotic events, in particular venous thromboembolism. Pulmonary embolism (PE) has been of special interest in patients with COVID-19 given its association with respiratory deterioration, increased risk of intensive care unit admission, and prolonged hospital stay. The pathophysiology and clinical characteristics of COVID-19-associated PE may differ from the conventional non-COVID-19-associated PE. In addition to embolic events from deep vein thrombi, in situ pulmonary thrombosis, particularly in smaller vascular beds, may be relevant in patients with COVID-19. Appropriate prevention of thrombotic events in COVID-19 has therefore become of critical interest. Several changes in viral biology, vaccination, and treatment management during the pandemic may have resulted in changes in incidence trends. This review provides an overview of the pathophysiology, epidemiology, clinical characteristics, and risk factors of COVID-19-associated PE. Furthermore, we briefly summarize the results from randomized controlled trials of preventive antithrombotic therapies in COVID-19, focusing on their findings related to PE. We discuss the acute treatment of COVID-19-associated PE, which is substantially similar to the management of conventional non-COVID-19 PE. Ultimately, we comment on the current knowledge gaps in the evidence and the future directions in the treatment and follow-up of COVID-19-associated PE, including long-term management, and its possible association with long-COVID.
Collapse
Affiliation(s)
- Luis Ortega-Paz
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida
| | - Azita H Talasaz
- Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Parham Sadeghipour
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
- Clinical Trial Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Tatjana S Potpara
- School of Medicine, University of Belgrade, Belgrade, Serbia
- Intensive Arrhythmia Care, Cardiology Clinic, Clinical Center of Serbia, Belgrade, Serbia
| | - Herbert D Aronow
- Department of Cardiology, Warren Alpert Medical School of Brown University, Providence, Rhode Island
- Department of Cardiology, Henry Ford Health, Detroit, Michigan
| | - Luis Jara-Palomares
- Respiratory Unit, Hospital Universitario Virgen del Rocio, Sevilla, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES), Carlos III Health Institute, Madrid, Spain
| | - Michelle Sholzberg
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Behnood Bikdeli
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Yale/YNHH Center for Outcomes Research and Evaluation (CORE), New Haven, Connecticut
- Cardiovascular Research Foundation (CRF), New York, New York
| |
Collapse
|
230
|
Casillas N, Ramón A, Torres AM, Blasco P, Mateo J. Predictive Model for Mortality in Severe COVID-19 Patients across the Six Pandemic Waves. Viruses 2023; 15:2184. [PMID: 38005862 PMCID: PMC10675561 DOI: 10.3390/v15112184] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/21/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
The impact of SARS-CoV-2 infection remains substantial on a global scale, despite widespread vaccination efforts, early therapeutic interventions, and an enhanced understanding of the disease's underlying mechanisms. At the same time, a significant number of patients continue to develop severe COVID-19, necessitating admission to intensive care units (ICUs). This study aimed to provide evidence concerning the most influential predictors of mortality among critically ill patients with severe COVID-19, employing machine learning (ML) techniques. To accomplish this, we conducted a retrospective multicenter investigation involving 684 patients with severe COVID-19, spanning from 1 June 2020 to 31 March 2023, wherein we scrutinized sociodemographic, clinical, and analytical data. These data were extracted from electronic health records. Out of the six supervised ML methods scrutinized, the extreme gradient boosting (XGB) method exhibited the highest balanced accuracy at 96.61%. The variables that exerted the greatest influence on mortality prediction encompassed ferritin, fibrinogen, D-dimer, platelet count, C-reactive protein (CRP), prothrombin time (PT), invasive mechanical ventilation (IMV), PaFi (PaO2/FiO2), lactate dehydrogenase (LDH), lymphocyte levels, activated partial thromboplastin time (aPTT), body mass index (BMI), creatinine, and age. These findings underscore XGB as a robust candidate for accurately classifying patients with COVID-19.
Collapse
Affiliation(s)
- Nazaret Casillas
- Department of Internal Medicine, Hospital Virgen De La Luz, 16002 Cuenca, Spain
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
| | - Antonio Ramón
- Department of Pharmacy, General University Hospital, 46014 Valencia, Spain
| | - Ana María Torres
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Pilar Blasco
- Department of Pharmacy, General University Hospital, 46014 Valencia, Spain
| | - Jorge Mateo
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| |
Collapse
|
231
|
Corsini CA, Filgueiras PS, Almeida NB, Miranda DAD, Gomes SV, Lourenço AJ, Bicalho CM, Assis JVD, Amorim RN, Silva RA, Vilela RV, Lima TM, Abreu DPD, Alvim RG, Castilho LR, Martins-Filho OA, Otta DA, Grenfell RF. Antibody response and soluble mediator profile in the first six months following acute SARS-CoV-2 infection. Sci Rep 2023; 13:18606. [PMID: 37903875 PMCID: PMC10616118 DOI: 10.1038/s41598-023-43263-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/21/2023] [Indexed: 11/01/2023] Open
Abstract
The COVID-19 pandemic has caused a severe global health and economic crisis, with significant consequences for human mortality and morbidity. Therefore, there is an urgent need for more studies on the immune response to SARS-CoV-2 infection, both to enhance its effectiveness and prevent its deleterious effects. This study presents the chronology of antibodies during six months after infection in hospitalized patients and the kinetics of serum soluble mediators of the cellular response triggered by SARS-CoV-2. Samples and clinical data from 330 patients hospitalized at the Hospital da Baleia in Belo Horizonte, Brazil, who were suspected of having COVID-19, were collected at the time of hospitalization and during 6 months after infection. The immune response was analyzed by enzyme-linked immunosorbent assay (ELISA) and flow cytometry. There was a significant difference in IgM specific antibody titers from the 7th to 60th days after infection between COVID-19 negative and positive patients. Soon after 60 days after infection, antibody levels started to reduce, becoming similar to the antibody levels of the COVID-19 negative patients. IgG specific antibodies started to be detectable after 9 days of infection and antibody levels were comparatively higher in positive patients as soon as after 7 days. Furthermore, IgG levels remained higher in these patients during the complete period of 180 days after infection. The study observed similar antibody profiles between different patient groups. The soluble systemic biomarkers evaluated showed a decrease during the six months after hospitalization, except for CCL11, CXCL8, CCL3, CCL4, CCL5, IL-6, IFN-g, IL-17, IL-5, FGF-basic, PDGF, VEGF, G-CSF, and GM-CSF. The results indicate that IgM antibodies are more prominent in the early stages of infection, while IgG antibodies persist for a longer period. Additionally, the study identified that patients with COVID-19 have elevated levels of biomarkers after symptom onset, which decrease over time.
Collapse
Affiliation(s)
- Camila A Corsini
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Priscilla S Filgueiras
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
- Department of Pathology, College of Medicine, Federal University of Minas Gerais, 6627 Avenida Presidente Antônio Carlos, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Nathalie Bf Almeida
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Daniel Ap de Miranda
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Sarah Vc Gomes
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Adelina Junia Lourenço
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
- Hospital da Baleia, Benjamin Guimarães Foundation, 1464 Juramento Street, Belo Horizonte, Minas Gerais, 30285-408, Brazil
| | - Cecilia Mf Bicalho
- Hospital da Baleia, Benjamin Guimarães Foundation, 1464 Juramento Street, Belo Horizonte, Minas Gerais, 30285-408, Brazil
| | - Jessica V de Assis
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
- Department of Pathology, College of Medicine, Federal University of Minas Gerais, 6627 Avenida Presidente Antônio Carlos, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Raquel Nh Amorim
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Raphael A Silva
- Hospital da Baleia, Benjamin Guimarães Foundation, 1464 Juramento Street, Belo Horizonte, Minas Gerais, 30285-408, Brazil
| | - Raquel Vr Vilela
- Hospital da Baleia, Benjamin Guimarães Foundation, 1464 Juramento Street, Belo Horizonte, Minas Gerais, 30285-408, Brazil
| | - Tulio M Lima
- Cell Culture Engineering Laboratory, COPPE, Universidade Federal do Rio de Janeiro, 550 Pedro Calmon Avenue, Rio de Janeiro, Rio de Janeiro, 21941-598, Brazil
| | - Daniel Pb de Abreu
- Cell Culture Engineering Laboratory, COPPE, Universidade Federal do Rio de Janeiro, 550 Pedro Calmon Avenue, Rio de Janeiro, Rio de Janeiro, 21941-598, Brazil
| | - Renata Gf Alvim
- Cell Culture Engineering Laboratory, COPPE, Universidade Federal do Rio de Janeiro, 550 Pedro Calmon Avenue, Rio de Janeiro, Rio de Janeiro, 21941-598, Brazil
| | - Leda R Castilho
- Cell Culture Engineering Laboratory, COPPE, Universidade Federal do Rio de Janeiro, 550 Pedro Calmon Avenue, Rio de Janeiro, Rio de Janeiro, 21941-598, Brazil
| | - Olindo A Martins-Filho
- Grupo Integrado de Pesquisa em Biomarcadores, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Dayane A Otta
- Grupo Integrado de Pesquisa em Biomarcadores, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Rafaella Fq Grenfell
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil.
- Department of Pathology, College of Medicine, Federal University of Minas Gerais, 6627 Avenida Presidente Antônio Carlos, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA, 30602-7387, USA.
| |
Collapse
|
232
|
Al Nafea HM, Al-Qahtani MT, Al Gahtani FH, Tabassum H. Blood coagulation, risk factors and associated complications in COVID-19 patients in Saudi Arabia: A retrospective cohort study. Medicine (Baltimore) 2023; 102:e35621. [PMID: 37904434 PMCID: PMC10615550 DOI: 10.1097/md.0000000000035621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/21/2023] [Indexed: 11/01/2023] Open
Abstract
A good understanding of the possible risk factors for coronavirus disease 19 (COVID-19) severity could help clinicians in identifying patients who need prioritized treatment to prevent disease progression and adverse outcomes. COVID-19-linked coagulopathy is one of the life-threatening severe acute respiratory syndrome coronavirus 2 infections. Growing evidence indicates a correlation between abnormal coagulation and increased risk of venous thromboembolism; in COVID-19-infected patients, yet a clear understanding of the role of coagulopathy in the severity of COVID-19 illness is still unresolved. This retrospective cohort study was thus undertaken to investigate the role of coagulation dysfunction with COVID-19 mortality/severity. Blood samples from 1000 hospitalized patients with COVID-19 pneumonia were collected. The study participants were both male and female in equal ratios with a mean age of 48.94. Patients were followed-up until discharge either for recovery or death. All biochemical investigations-complete blood count and coagulation profile including D-dimers, prothrombin time, partial prothrombin time, and international normalized ratio was performed in COVID-19 survivors and in non-survivors admitted in intensive care unit. In the survivor group, all coagulation parameters were within normal limits, and 8.7% had a low red blood count. The most common risk factors associated with COVID-19 patients were diabetes mellitus (2.8%), hypertension (10.8%), and heart disease (3%). In the non-survivor group, the coagulation parameters were above the normal range (prothrombin in 31.5%, PTT in 10.5%, international normalized ratio in 26.3%, D-dimer in 36.8%) with thrombocytopenia in 21.04% of patients. Other complications were pulmonary embolism in 21.05% and venous thromboembolism in 15.7% of non-survivors. A significant association was found between increased markers of coagulopathy and the severity of SARS-CoV2 infection. Furthermore, the severity of infection was observed to increase with risk factors such as age, heart disease, hypertension, and DM eventually affecting COVID-19 prognosis and mortality.
Collapse
Affiliation(s)
- Haifa Mohammed Al Nafea
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Tahani Al-Qahtani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Farjah Hassan Al Gahtani
- Department of Medicine, Division of Hematology/Oncology (Oncology Center), College of Medicine, King Khalid University Hospital, King Saud University, Riyadh, Saudi Arabia
| | - Hajera Tabassum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
233
|
Figueirêdo Leite GG, Colo Brunialti MK, Peçanha-Pietrobom PM, Abrão Ferreira PR, Ota-Arakaki JS, Cunha-Neto E, Ferreira BL, Ronsein GE, Tashima AK, Salomão R. Understanding COVID-19 progression with longitudinal peripheral blood mononuclear cell proteomics: Changes in the cellular proteome over time. iScience 2023; 26:107824. [PMID: 37736053 PMCID: PMC10509719 DOI: 10.1016/j.isci.2023.107824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/16/2023] [Accepted: 08/31/2023] [Indexed: 09/23/2023] Open
Abstract
The clinical presentation of COVID-19 is highly variable, and understanding the underlying biological processes is crucial. This study utilized a proteomic analysis to investigate dysregulated processes in the peripheral blood mononuclear cells of patients with COVID-19 compared to healthy volunteers. Samples were collected at different stages of the disease, including hospital admission, after 7 days of hospitalization, and 30 days after discharge. Metabolic pathway alterations and increased abundance of neutrophil-related proteins were observed in patients. Patients progressing to critical illness had significantly low-abundance proteins in the pentose phosphate and glycolysis pathways compared with those presenting clinical recovery. Important biological processes, such as fatty acid concentration and glucose metabolism disorder, remained altered even after 30 days of hospital discharge. Temporal proteomic changes revealed distinct pathways in critically ill and non-critically ill patients. Our study emphasizes the significance of longitudinal cellular proteomic studies in identifying disease progression-related pathways and persistent protein changes post-hospitalization.
Collapse
Affiliation(s)
| | - Milena Karina Colo Brunialti
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Paula M. Peçanha-Pietrobom
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Paulo R. Abrão Ferreira
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Jaquelina Sonoe Ota-Arakaki
- Division of Respiratory Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Bianca Lima Ferreira
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Graziella E. Ronsein
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP, Brazil
| | - Alexandre Keiji Tashima
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Reinaldo Salomão
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
234
|
Amalia L. Correlation Between Hypercoagulable State and Severity Level of Ischemic Stroke With Covid-19 Infection. J Blood Med 2023; 14:537-542. [PMID: 37849564 PMCID: PMC10577248 DOI: 10.2147/jbm.s429357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/06/2023] [Indexed: 10/19/2023] Open
Abstract
Background Hypercoagulable state in acute ischemic stroke patients with COVID-19, was found to occur in most cases, may affect the severity and clinical outcome of acute ischemic stroke with COVID-19. Ischemic stroke patients with COVID-19 infection have worsen prognosis in mortality regarding hypercoagulable state condition. Objective The study aims to determine the relationship between the hypercoagulable state and the severity of acute ischemic stroke patients with COVID-19. Methods This study is a retrospective analytic study using a cross-sectional method in acute ischemic stroke who meet the criteria must have focal clinical symptoms or global dysfunction lasting more than 24 hours, be caused by vascular factors, be confirmed positive for COVID-19, NIHSS (admission and discharge), and have an examination of D-dimer and/or fibrinogen. Chi-Square is used for data processing relationship analysis. Results A total of 32 patients met the inclusion and exclusion criteria of this study. Elevated D-dimer and/or fibrinogen were found in 28 patients (87.5%), confirming a hypercoagulable state. In this study, the average value of D-dimer was 5.3 mg/mL, and fibrinogen was 479 mg/dL. Based on the admission NIHSS score, it was found that most of the patients had moderate strokes with an average NIHSS score of 12. The chi-square test results showed no relationship between the hypercoagulable state and the severity of acute ischemic stroke as measured by NIHSS admission (p=0.333), but it was closely related to NIHSS exit (p=0.02). The finding supports that 40.62% of acute ischemic stroke patients with COVID-19 confirmed to have a hypercoagulable state had a death discharge status. Conclusion There is no significant relationship between hypercoagulable state and stroke severity on admission, but it closely related to NIHSS on discharge and high mortality in acute ischemic stroke patients with COVID-19.
Collapse
Affiliation(s)
- Lisda Amalia
- Department of Neurology, Medical Faculty, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
235
|
Făgărășan I, Rusu A, Comșa H, Simu TD, Vulturar DM, Todea DA. IL-6 and Neutrophil/Lymphocyte Ratio as Markers of ICU Admittance in SARS-CoV-2 Patients with Diabetes. Int J Mol Sci 2023; 24:14908. [PMID: 37834356 PMCID: PMC10573809 DOI: 10.3390/ijms241914908] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/18/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Inflammation along with coagulation disturbances has an essential role in the evolution towards a severe disease in patients with the coronavirus disease 2019 (COVID-19). This study aimed to evaluate inflammatory and coagulation biomarkers when predicting the need to visit an intensive care unit (ICU) in diabetes mellitus (DM) patients. In a retrospective study, laboratory parameters were examined for 366 participants: ICU = 90, of which 44 patients had DM and no ICU admittance = 276. The ability of inflammatory and coagulation markers to distinguish the severity of COVID-19 was determined using univariate and multivariate regression analysis. In all patients, lactate dehydrogenase was the only predictor for ICU admittance in the multivariate analysis. In the DM group, the results showed that the interleukin (IL)-6 and neutrophil/lymphocyte ratio (NLR) values at admission could predict the need for ICU admittance. Even though there were significant differences between the ICU and no ICU admittance groups regarding the coagulation markers, they could not predict the severity of the disease in DM patients. The present study showed for the first time that the IL-6 and NLR admission values could predict ICU admittance in DM patients. This finding could help clinicians manage the infection more easily if the COVID-19 pandemic strikes again.
Collapse
Affiliation(s)
- Iulia Făgărășan
- Department of Pneumology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania; (I.F.); (D.-M.V.); (D.-A.T.)
| | - Adriana Rusu
- Department of Diabetes and Nutrition Diseases, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Horațiu Comșa
- Cardiology Department, Clinical Rehabilitation Hospital, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Tudor-Dan Simu
- Intensive Care Department, “Leon Daniello” Pulmonology Hospital, 400332 Cluj-Napoca, Romania;
| | - Damiana-Maria Vulturar
- Department of Pneumology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania; (I.F.); (D.-M.V.); (D.-A.T.)
| | - Doina-Adina Todea
- Department of Pneumology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania; (I.F.); (D.-M.V.); (D.-A.T.)
| |
Collapse
|
236
|
Walborn AT, Heath A, Neal MD, Zarychanski R, Kornblith LZ, Hunt BJ, Castellucci LA, Hochman JS, Lawler PR, Paul JD. Effects of inflammation on thrombosis and outcomes in COVID-19: secondary analysis of the ATTACC/ACTIV-4a trial. Res Pract Thromb Haemost 2023; 7:102203. [PMID: 37854455 PMCID: PMC10579532 DOI: 10.1016/j.rpth.2023.102203] [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: 11/22/2022] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 10/20/2023] Open
Abstract
Background Patients hospitalized for COVID-19 are at high risk of thrombotic complications and organ failure, and often exhibit severe inflammation, which may contribute to hypercoagulability. Objectives To determine whether patients hospitalized for COVID-19 experience differing frequencies of thrombotic and organ failure complications and derive variable benefits from therapeutic-dose heparin dependent on the extent of systemic inflammation and whether observed benefit from therapeutic-dose anticoagulation varies depending on the degree of systemic inflammation. Methods We analyzed data from 1346 patients hospitalized for COVID-19 enrolled in the ATTACC and ACTIV-4a platforms who were randomized to therapeutic-dose heparin or usual care for whom levels of C-reactive protein (CRP) were reported at baseline. Results Increased CRP was associated with worse patient outcomes, including a >98% posterior probability of increased organ support requirement, hospital length of stay, risk of 28-day mortality, and incidence of major thrombotic events or death (patients with CRP 40-100 mg/L or ≥100 mg/L compared to patients with CRP <40 mg/L). Patients with CRP 40 to 100 mg/L experienced the greatest degree of benefit from treatment with therapeutic doses of unfractionated or low molecular weight heparin compared with usual-care prophylactic doses. This was most significant for an increase in organ support-free days (odds ratio: 1.63; 95% confidence interval, 1.09-2.40; 97.9% posterior probability of beneficial effect), with trends toward benefit for other evaluated outcomes. Conclusion Moderately ill patients hospitalized for COVID-19 with CRP between 40 mg/L and 100 mg/L derived the greatest benefit from treatment with therapeutic-dose heparin.
Collapse
Affiliation(s)
- Amanda T. Walborn
- Department of Anesthesia and Critical Care, University of Chicago Medical Center, Chicago, Illinois, USA
| | - Anna Heath
- The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of the Biostatistics, The University of Toronto, Toronto, Ontario, Canada
- Department of Statistical Science, University College London, London, UK
| | - Matthew D. Neal
- Pittsburgh Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ryan Zarychanski
- Department of Internal Medicine, Sections of Hematology/Medical Oncology and Critical Care, Max Rad College of Medicine, University of Manitoba, Winnipeg, Manitoba
| | - Lucy Z. Kornblith
- University of California, San Francisco, San Francisco, California, USA
| | - Beverley J. Hunt
- Thrombosis & Haemophilia Centre, Kings Healthcare Partners, London, UK
| | - Lana A. Castellucci
- Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Judith S. Hochman
- Department of Medicine, Section of Cardiology, NYU Langone Health, New York, New York, USA
| | - Patrick R. Lawler
- Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, Ontario, Canada
- Division of Cardiology and Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan D. Paul
- Department of Medicine, Section of Cardiology, University of Chicago Medical Center, Chicago, Illinois, USA
| |
Collapse
|
237
|
Mitroi RM, Padureanu V, Mitrea A, Protasiewicz Timofticiuc DC, Rosu MM, Clenciu D, Enescu A, Padureanu R, Tenea Cojan TS, Vladu IM. Prothrombotic status in COVID‑19 with diabetes mellitus (Review). Biomed Rep 2023; 19:65. [PMID: 37649534 PMCID: PMC10463232 DOI: 10.3892/br.2023.1647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 07/26/2023] [Indexed: 09/01/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has caused an important social and health impact worldwide and the coronavirus disease-19 (COVID-19) has elicited devastating economy problems. The pathogenesis of SARS-CoV-2 infection is a complex mechanism and is considered to be the result of a challenging interaction, in which host and virus immune responses are the key elements. In this process, several inflammatory pathways are involved, and their initiation can have multiple consequences with a considerable impact on evolution, such as hyperinflammation and cytokine storm, thereby promoting activation of the coagulation system and fibrinolytic activity suppression. It is commonly recognized that COVID-19 severity involves multiple factors, including diabetes which increases the risk of developing different complications. This could be as a result of the low-grade inflammation as well as the innate and adaptive immune response dysfunction that is observed in patients with diabetes mellitus. In patients with diabetes, multiple metabolic disturbances which have a major impact in disturbing the balance between coagulation and fibrinolysis were discovered, thus the risk for thrombotic events is increased. Diabetes has been recognized as an important severity prognosis factor in COVID-19 cases and considering there is a significant association between diabetes and prothrombotic status, it could be responsible for the increased risk of thrombotic events with a worse prognosis.
Collapse
Affiliation(s)
- Roxana Madalina Mitroi
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Vlad Padureanu
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Adina Mitrea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Diabetes, Nutrition and Metabolic Diseases, Clinical Municipal Hospital ‘Philanthropy’ of Craiova, 200143 Craiova, Romania
| | | | - Maria Magdalena Rosu
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Diana Clenciu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Diabetes, Nutrition and Metabolic Diseases, Clinical Municipal Hospital ‘Philanthropy’ of Craiova, 200143 Craiova, Romania
| | - Aurelia Enescu
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Rodica Padureanu
- Department of Pneumology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Tiberiu Stefanita Tenea Cojan
- Department of General Surgery, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ionela Mihaela Vladu
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
238
|
Montorfano M, Leoni O, Andreassi A, Ludergnani M, Moroni F, Ancona MB, Landoni G, Ciceri F, Zangrillo A. Chronic anticoagulant treatment and risk of mortality in SARS-Cov2 patients: a large population-based study. Minerva Med 2023; 114:628-633. [PMID: 35191294 DOI: 10.23736/s0026-4806.22.07797-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Hypercoagulability is often seen in COVID-19 patients and thromboembolic events appear frequent; antithrombotic treatment has been proposed therefore as part of standard treatment for COVID-19. Under these premises, prior-to-infection antithrombotic treatment may have a protective effect with respect to COVID-19 related thromboembolic events. Aim of the present work was to evaluate the impact of prior-to-infection anticoagulant or antiplatelet treatment on COVID-19 outcomes. METHODS Beneficiaries of the Regional Health Service of the Lombardy region of Italy aged ≥40 years with a COVID-19 diagnosis made between February 21st and July 18th, 2020 were included in the present study. The impact on COVID-19 mortality of pre-existing and chronic therapy with anticoagulant drugs (vitamin-K antagonist or new oral anticoagulants) was evaluated. Analyses were repeated with antiplatelets drugs. RESULTS Among 79,934 SARS-CoV-2 patients beneficiaries of the Regional Healthcare System of the Lombardy Region who received a diagnosis between February 21st and July 18th, 2020, chronic pre-existing anticoagulant assumption was present in 6.0% and antiplatelets in 12.7%. The overall unadjusted mortality rate was 20.6%, with male sex, age category and comorbidity burden being significantly associated to increased mortality risk. Anticoagulant chronic treatment was not associated with a reduction in mortality. Similar results were observed when repeating the analyses for pre-existing oral antiplatelet treatment. CONCLUSIONS In a large population-based study evaluating more than 79,000 COVID-19 patients, pre-existing antithrombotic therapy was not associated to a benefit in terms of mortality. Further studies are needed to evaluate the role of antithrombotic therapy as standard treatment among COVID-19 patients.
Collapse
Affiliation(s)
- Matteo Montorfano
- Unit of Interventional Cardiology, IRCCS San Raffaele Scientific Institute, Milan, Italy -
| | - Olivia Leoni
- Welfare Directorate, Regione Lombardia, Milan, Italy
| | | | | | - Francesco Moroni
- Unit of Interventional Cardiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco B Ancona
- Unit of Interventional Cardiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Hematology and Bone Marrow Transplant, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Zangrillo
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
239
|
Busuioc CJ, Roşu GC, Zorilă GL, Mogoantă L, Istrate-Ofiţeru AM, Pirici D, Liliac IM, Iovan L, Berbecaru EIA, Comănescu MC, Cazacu SM, Iliescu DG. The influence of SARS-CoV-2 on the immune system elements and on the placental structure. Clinical, histological and immunohistochemical study. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2023; 64:549-557. [PMID: 38184836 PMCID: PMC10863699 DOI: 10.47162/rjme.64.4.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND The effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during pregnancy remain relatively unknown. AIM We present this original paper where we analyzed 60 parturients, at term, 30 without associated infection (C-) and 30 with associated infection (C+), present at birth. METHODS We analyzed the blood count and placental microscopic structure through classical and immunohistochemical staining and observed the placental areas affected by the presence of SARS-CoV-2. RESULTS SARS-CoV-2 infection was accompanied by a decrease in the number of lymphocytes, the number of platelets and the presence of placental structural changes, identifying extensive areas of amyloid deposits, placental infarcts, vascular thrombosis, syncytial knots, with a decrease in placental vascular density and the presence of infection in the cells located at decidual level, at syncytiotrophoblast level and at the level of the cells of the chorionic plate, still without overcoming this barrier and without causing any fetal infection in the analyzed cases. CONCLUSIONS This study shows that the invasion of SARS-CoV-2 in the placenta can produce significant structural changes, with a decrease in placental vascular density that can have significant implications on proper fetal perfusion. Also, the presence of immunoreactivity at the level of decidua, the placental villi, as well as the chorionic plate proves that the virus can overcome the maternal-fetal barrier. However, in the analyzed cases there were no fetal infections at birth, which may show that local placental factors can be a protective filter for the fetus.
Collapse
Affiliation(s)
| | | | - George-Lucian Zorilă
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy of Craiova, Romania
| | - Laurenţiu Mogoantă
- Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
- Romanian Academy of Medical Sciences, Craiova Subsidiary, Romania
| | | | - Daniel Pirici
- Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| | - Ilona Mihaela Liliac
- Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| | - Larisa Iovan
- Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| | | | | | - Sergiu Marian Cazacu
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, Romania
| | - Dominic-Gabriel Iliescu
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy of Craiova, Romania
| |
Collapse
|
240
|
Kroll MH, Bi C, Salm AE, Szymanski J, Goldstein DY, Wolgast LR, Rosenblatt G, Fox AS, Kapoor H. Risk Estimation of Severe COVID-19 Based on Initial Biomarker Assessment Across Racial and Ethnic Groups. Arch Pathol Lab Med 2023; 147:1109-1118. [PMID: 37338199 DOI: 10.5858/arpa.2023-0039-sa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2023] [Indexed: 06/21/2023]
Abstract
CONTEXT.— Disease courses in COVID-19 patients vary widely. Prediction of disease severity on initial diagnosis would aid appropriate therapy, but few studies include data from initial diagnosis. OBJECTIVE.— To develop predictive models of COVID-19 severity based on demographic, clinical, and laboratory data collected at initial patient contact after diagnosis of COVID-19. DESIGN.— We studied demographic data and clinical laboratory biomarkers at time of diagnosis, using backward logistic regression modeling to determine severe and mild outcomes. We used deidentified data from 14 147 patients who were diagnosed with COVID-19 by polymerase chain reaction SARS-CoV-2 testing at Montefiore Health System, from March 2020 to September 2021. We generated models predicting severe disease (death or more than 90 hospital days) versus mild disease (alive and fewer than 2 hospital days), starting with 58 variables, by backward stepwise logistic regression. RESULTS.— Of the 14 147 patients, including Whites, Blacks, and Hispanics, 2546 (18%) patients had severe outcomes and 3395 (24%) had mild outcomes. The final number of patients per model varied from 445 to 755 because not all patients had all available variables. Four models (inclusive, receiver operating characteristic, specific, and sensitive) were identified as proficient in predicting patient outcomes. The parameters that remained in all models were age, albumin, diastolic blood pressure, ferritin, lactic dehydrogenase, socioeconomic status, procalcitonin, B-type natriuretic peptide, and platelet count. CONCLUSIONS.— These findings suggest that the biomarkers found within the specific and sensitive models would be most useful to health care providers on their initial severity evaluation of COVID-19.
Collapse
Affiliation(s)
- Martin H Kroll
- From the Department of Medical Operations and Quality (Kroll), Quest Diagnostics, Secaucus, New Jersey
| | - Caixia Bi
- Department of Corporate Medical (Bi), Quest Diagnostics, Secaucus, New Jersey
| | - Ann E Salm
- Department of Infectious Diseases/Immunology (Salm, Kapoor), Quest Diagnostics, Secaucus, New Jersey
| | - James Szymanski
- Department of Pathology, Montefiore Medical Center, Bronx, New York (Szymanski, Goldstein, Wolgast, Fox)
| | - D Yitzchak Goldstein
- Department of Pathology, Montefiore Medical Center, Bronx, New York (Szymanski, Goldstein, Wolgast, Fox)
| | - Lucia R Wolgast
- Department of Pathology, Montefiore Medical Center, Bronx, New York (Szymanski, Goldstein, Wolgast, Fox)
| | - Gregory Rosenblatt
- The Department of Pathology, Albert Einstein College of Medicine, Bronx, New York (Rosenblatt). Kapoor is currently located at HK Healthcare Consultant LLC in Davie, Florida
| | - Amy S Fox
- Department of Pathology, Montefiore Medical Center, Bronx, New York (Szymanski, Goldstein, Wolgast, Fox)
| | - Hema Kapoor
- Department of Infectious Diseases/Immunology (Salm, Kapoor), Quest Diagnostics, Secaucus, New Jersey
| |
Collapse
|
241
|
Baptista A, Vieira AM, Capela E, Julião P, Macedo A. COVID-19 fatality rates in hospitalized patients: A new systematic review and meta-analysis. J Infect Public Health 2023; 16:1606-1612. [PMID: 37579698 DOI: 10.1016/j.jiph.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/21/2023] [Accepted: 07/14/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND SARS-COV2 or COVID-19 disease is an infectious illness that emerged for the first time at the end of 2019, in Wuhan, China and rapidly turned out to be an international pandemic with deleterious effects all over the world. In March 2021, A. Macedo et al., has published the first meta-analysis of hospital mortality, so the authors decided to update those data at a time of emergence of new therapies and increasing vaccination rates. METHODS As the outcome of interest was the mortality in hospitalized general patients, the authors looked for articles evaluating the clinical characteristics of those patients, consulting PUBMED (The US National Library of Medicine) and EMBASE (Medical database) in an independent selection using predefined terms of search. A meta-analysis random-effect model was estimated using Mantel-Haenszel method. Heterogeneity among studies was tested using Tau2 statistics and Chi2 statistics. RESULTS In a first instance 25 articles were included for final analysis with a total of 103,840 patients, but as the goal was to update the anterior data, these studies were analysed together with the 21 studies of the previous meta-analysis, with a total of 114609 patients. The mortality rate of COVID-19 general patients admitted to the hospital was 16% (95% CI 12; 21, I2 =100%). CONCLUSION Global hospital mortality of COVID-19 of general patients was 16%, with quite different rates according to the different geographic areas analysed.
Collapse
Affiliation(s)
- Alexandre Baptista
- Faculdade de Medicina e Ciências Biomédicas Universidade Algarve, Faro, Portugal
| | - Ana M Vieira
- Faculdade de Medicina e Ciências Biomédicas Universidade Algarve, Faro, Portugal
| | - Eunice Capela
- Faculdade de Medicina e Ciências Biomédicas Universidade Algarve, Faro, Portugal
| | - Pedro Julião
- Faculdade de Medicina e Ciências Biomédicas Universidade Algarve, Faro, Portugal
| | - Ana Macedo
- Faculdade de Medicina e Ciências Biomédicas Universidade Algarve, Faro, Portugal; Algarve Biomedical Center, Faro, Portugal.
| |
Collapse
|
242
|
Khoramipour M, Jalali A, Abbasi B, Hadi Abbasian M. Evaluation of the association between clinical parameters and ADAM33 and ORMDL3 asthma gene single-nucleotide polymorphisms with the severity of COVID-19. Int Immunopharmacol 2023; 123:110707. [PMID: 37499392 DOI: 10.1016/j.intimp.2023.110707] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Coronavirus Disease of 2019 (COVID-19) is a contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Patients had varying clinical symptoms and disease severity (mild, moderate, severe, and critical). Several risk factors, including genetic polymorphisms, have been reported to be associated with disease risk and severity. This study aimed to investigate the association of two polymorphisms in the orosomucoid1-like 3 (ORMDL3) and a disintegrin and metalloprotease 33 (ADAM33) asthma-related genes with the severity of COVID-19. MATERIAL AND METHODS The study included 116 COVID-19 patients with a positive polymerase chain reaction (PCR) test for the SARS-CoV-2 Delta variant. 58 patients with moderate symptoms, 28 patients with severe symptoms, and 30 outpatients with mild symptoms. Genotyping of rs7216389 in the ORMDL3 and rs2280091 in ADAM33 genes was performed by polymerase chain reaction-restriction fragment length polymorphism. Furthermore, records of patients were studied for hematological profiles and biochemical markers. RESULTS No significant association was found between rs7216389 and rs2280091 and the severity of COVID-19 between different groups of COVID-19 patients. The serum levels of RBC and neutrophil-to-lymphocyte ratio were significantly increased; the erythrocyte sedimentation rate (ESR), and Aspartate transaminase (SGOT) were significantly decreased during treatment in intensive care unit (ICU) patients. The serum levels of red blood cells, Platelets, Urea, Alkaline phosphatase, ESR, Alanine transaminase (SGPT), and SGOT were significantly increased during treatment in hospitalized patients. The serum levels of inflammatory factors, including C-reactive protein (CRP), D-dimer, and Ferritin at the time of admission, were significantly higher in patients admitted to the ICU patients compared to the other group of patients. CONCLUSION The two polymorphisms studied in this research are not suitable markers for predicting the severity of COVID-19. However, there are significant differences in the amounts of some blood factors in different groups of COVID-19 patients (P < 0.05) and these factors can be used as a marker for the disease severity prediction.
Collapse
Affiliation(s)
- Mahsa Khoramipour
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran
| | - Amir Jalali
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran.
| | - Bahareh Abbasi
- Department of Medical Genetics, National Institute for Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mohammad Hadi Abbasian
- Department of Medical Genetics, National Institute for Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
243
|
Hao W, Liu M, Bai C, Liu X, Niu S, Chen X. Increased inflammatory mediators levels are associated with clinical outcomes and prolonged illness in severe COVID-19 patients. Int Immunopharmacol 2023; 123:110762. [PMID: 37562295 DOI: 10.1016/j.intimp.2023.110762] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023]
Abstract
OBJECTIVE The purpose of this study was to identify potential predictors of clinical outcome in severe COVID-19 patients and to investigate the relationship between immunological parameters and duration of illness. METHODS This single-center study retrospectively recruited 73 patients with severe or critical COVID-19. Immunological indicators include white blood cell count, neutrophil count, lymphocyte count, neutrophil-to-lymphocyte ratio, and circulating inflammatory mediators were observed for their association with disease severity, mortality and duration of illness of COVID-19. RESULTS Serum inflammatory mediators levels of C-reactive protein (P = 0.015), interleukin 6 (IL-6) (P < 0.001), CX3CL1 (P < 0.001), D-dimer (P < 0.001) and procalcitonin (PCT) (P < 0.001) were increased in critical illness patients compared to those severe COVID-19 patients. CX3CL1 has the highest C-index (0.75) to predict in-hospital mortality in patients with COVID-19. Furthermore, this study shows for the first time that the duration of illness in severe COVID-19 patients is associated with serum levels of CX3CL1 (P = 0.037) and D-dimer (P = 0.014). CONCLUSION CX3CL1, D-dimer, PCT, and IL-6 could effectively predict mortality in severe COVID-19 patients. In addition, only the circulating levels of CX3CL1 and D-dimer were significantly associated with duration of illness.
Collapse
Affiliation(s)
- Wendong Hao
- Department of Allergy, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China; Department of Respiratory and Critical Care Medicine, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China.
| | - Meimei Liu
- Department of Allergy, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China; Department of Respiratory and Critical Care Medicine, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China
| | - Cairong Bai
- Department of Allergy, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China
| | - Xin Liu
- Department of Allergy, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China; Department of Respiratory and Critical Care Medicine, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China
| | - Siqian Niu
- Department of Respiratory and Critical Care Medicine, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China
| | - Xiushan Chen
- Department of Respiratory and Critical Care Medicine, Yulin Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Yulin 719000, Shaanxi province, PR China
| |
Collapse
|
244
|
Howick V JF, Harmon DM, McBane RD. 44-Year-Old Woman With Cough and Shortness of Breath. Mayo Clin Proc 2023; 98:1557-1563. [PMID: 37793730 DOI: 10.1016/j.mayocp.2023.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 10/06/2023]
Affiliation(s)
- James F Howick V
- Resident in Internal Medicine, Mayo Clinic School of Graduate Medical Education, Rochester, MN
| | - David M Harmon
- Resident in Internal Medicine, Mayo Clinic School of Graduate Medical Education, Rochester, MN
| | - Robert D McBane
- Advisor to residents and Consultant in Vascular Cardiology, Mayo Clinic, Rochester, MN.
| |
Collapse
|
245
|
Neves FF, Pott-Junior H, Yamashita KMC, de Sousa Santos S, Cominetti MR, de Melo Freire CC, Cunha AFD, Jordão Júnior AA. Do the oxidative stress biomarkers predict COVID-19 outcome? An in-hospital cohort study. Free Radic Biol Med 2023; 207:194-199. [PMID: 37454917 DOI: 10.1016/j.freeradbiomed.2023.06.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/07/2023] [Accepted: 06/24/2023] [Indexed: 07/18/2023]
Abstract
In SARSCoV-2 infections, excessive activation of the immune system dramatically elevates reactive oxygen species levels, harms cell structures, and directly increases disease severity and mortality. We aimed to evaluate whether plasma oxidative stress biomarker levels could predict mortality in adults admitted with Coronavirus Disease 2019 (COVID-19), considering potential confounders. We conducted a cohort study of 115 adults (62.1 ± 17.6 years, 65 males) admitted to a Brazilian public hospital for severely symptomatic COVID-19. Serum levels of α-tocopherol, glutathione, superoxide dismutase, 8-hydroxy-2'-deoxyguanosine, malondialdehyde, and advanced oxidation protein products were quantified at COVID-19 diagnosis using real-time polymerase chain reaction. Serum levels of α-tocopherol, glutathione, superoxide dismutase, and advanced oxidation protein products differed significantly between survivors and non-survivors. Serum glutathione levels below 327.2 μmol/mL were associated with a significant risk of death in COVID-19 patients, even after accounting for other factors (adjusted hazard ratio = 3.12 [95% CI: 1.83-5.33]).
Collapse
Affiliation(s)
- Fabio Fernandes Neves
- Department of Medicine, Federal University of São Carlos - UFSCar, 13565-905, São, Carlos, Brazil.
| | - Henrique Pott-Junior
- Department of Medicine, Federal University of São Carlos - UFSCar, 13565-905, São, Carlos, Brazil
| | | | - Sigrid de Sousa Santos
- Department of Medicine, Federal University of São Carlos - UFSCar, 13565-905, São, Carlos, Brazil
| | - Marcia Regina Cominetti
- Department of Gerontology, Federal University of São Carlos - UFSCar, 13565-905, São, Carlos, Brazil
| | - Caio Cesar de Melo Freire
- Department of Genetics and Evolution, Federal University of São Carlos - UFSCar, 13565-905, São Carlos, Brazil
| | - Anderson Ferreira da Cunha
- Department of Genetics and Evolution, Federal University of São Carlos - UFSCar, 13565-905, São Carlos, Brazil
| | - Alceu Afonso Jordão Júnior
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo - USP, 14049-900, Ribeirão Preto, Brazil
| |
Collapse
|
246
|
Thaler S, Stöhr D, Kammerer T, Nitschke T, Hoechter DJ, Brandes F, Müller M, Groene P, Schäfer ST. Predictive value of coagulation variables and glycocalyx shedding in hospitalized COVID-19 patients - a prospective observational study. Acta Clin Belg 2023; 78:392-400. [PMID: 37092324 DOI: 10.1080/17843286.2023.2204593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/15/2023] [Indexed: 04/25/2023]
Abstract
OBJECTIVES Covid-19 disease causes an immense burden on the healthcare system. It has not yet been finally clarified which patients will suffer from a severe course and which will not. Coagulation disorders can be detected in many of these patients. The aim of the present study was therefore to identify variables of the coagulation system including standard and viscoelastometric tests as well as components of glycocalyx damage that predict admission to the intensive care unit. METHODS Adult patients were included within 24 h of admission. Blood samples were analyzed at hospital admission and at ICU admission if applicable. We analyzed group differences and furthermore performed receiver operator characteristics (ROC). RESULTS This study included 60 adult COVID-19 patients. During their hospital stay, 14 patients required ICU treatment. Comparing ICU and non-ICU patients at time of hospital admission, D-dimer (1450 µg/ml (675/2850) vs. 600 µg/ml (500/900); p = 0.0022; cut-off 1050 µg/ml, sensitivity 71%, specificity 89%) and IL-6 (47.6 pg/ml (24.9/85.4 l) vs. 16.1 pg/ml (5.5/34.4); p = 0.0003; cut-off 21.25 pg/ml, sensitivity 86%, specificity 65%) as well as c-reactive protein (92 mg/dl (66.8/131.5) vs. 43.5 mg/dl (26.8/83.3); p = 0.0029; cutoff 54.5 mg/dl, sensitivity 86%, specificity 65%) were higher in patients who required ICU admission. Thromboelastometric variables and markers of glycocalyx damage (heparan sulfate, hyaluronic acid, syndecan-1) at the time of hospital admission did not differ between groups. CONCLUSION General inflammatory variables continue to be the most robust predictors of a severe course of a COVID-19 infection. Viscoelastometric variables and markers of glycocalyx damage are significantly increased upon admission to the ICU without being predictors of ICU admission.
Collapse
Affiliation(s)
- Sarah Thaler
- Department of Anaesthesiology, University Hospital, Munich, Germany
| | - Dana Stöhr
- Department of Anaesthesiology, University Hospital, Munich, Germany
| | - Tobias Kammerer
- Department of Anaesthesiology, University Hospital, Munich, Germany
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany
| | - Tobias Nitschke
- Department of Anaesthesiology, University Hospital, Munich, Germany
| | | | - Florian Brandes
- Department of Anaesthesiology, University Hospital, Munich, Germany
| | - Martin Müller
- Department of Anaesthesiology, University Hospital, Munich, Germany
| | - Philipp Groene
- Department of Anaesthesiology, University Hospital, Munich, Germany
| | - Simon T Schäfer
- Department of Anaesthesiology, University Hospital, Munich, Germany
| |
Collapse
|
247
|
Bentounes NK, Chocron R, Philippe A, Smadja DM, Gendron N. Impact of COVID-19 Pandemic on Temporal Trends of Hemostasis Test in France: A Retrospective Analysis of 9 Years of National Health Data. TH OPEN 2023; 7:e285-e288. [PMID: 37818324 PMCID: PMC10562010 DOI: 10.1055/a-2165-1249] [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: 07/19/2023] [Accepted: 08/31/2023] [Indexed: 10/12/2023] Open
Affiliation(s)
- Nûn K. Bentounes
- University Paris Cité, Innovative Therapies in Haemostasis, INSERM, Paris, France
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris. Centre-Université de Paris, Paris, France
| | - Richard Chocron
- Emergency Department, Assistance Publique Hôpitaux de Paris. Centre-Université de Paris, Paris, France
- University Paris Cité, Paris Cardiovascular Research Center (PARCC), INSERM, Paris, France
| | - Aurélien Philippe
- University Paris Cité, Innovative Therapies in Haemostasis, INSERM, Paris, France
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris. Centre-Université de Paris, Paris, France
| | - David M. Smadja
- University Paris Cité, Innovative Therapies in Haemostasis, INSERM, Paris, France
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris. Centre-Université de Paris, Paris, France
- French Clinical Research Infrastructure Network (F-CRIN), Investigation Network On Venous Thrombo-Embolism (INNOVTE), Saint-Étienne, France
| | - Nicolas Gendron
- University Paris Cité, Innovative Therapies in Haemostasis, INSERM, Paris, France
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris. Centre-Université de Paris, Paris, France
| |
Collapse
|
248
|
Cicco S, Vacca A, Albanese F, Susca N, Desantis V, Magistro A, Cazzato G, Cicco G, Sablone S, Cariddi C, Marozzi MS, Catena C, Brosolo G, Marcante S, Ingravallo G, Dalfino L, Lauletta G, Pappagallo F, Solimando AG, Grasso S, Maiorano E, Introna F, Sechi LA, Ria R. Immune disturbance leads to pulmonary embolism in COVID-19 more than classical risk factors: a clinical and histological study. Intern Emerg Med 2023; 18:1981-1993. [PMID: 37592135 PMCID: PMC10543807 DOI: 10.1007/s11739-023-03383-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 07/19/2023] [Indexed: 08/19/2023]
Abstract
COVID-19 induces endotheliitis and one of the main complications is enhanced coagulation. The incidence of pulmonary embolism (PE) in COVID-19 (CPE) has increased and clinical features for a rigorous analysis still need to be determined. Thus, we evaluated the clinical characteristics in CPE and the immune infiltration that occurred. Between January 1 and December 31, 2021, 38 patients were affected by CPE (9 ICU, 19 males/19 females, 70.18 ± 11.24 years) out of 459 COVID-19 cases. Controls were subjects who were evaluated for PE between January 1 2015, and December 31, 2019 (92 patients, 9 ICU, 48 males/45 females, 69.55 ± 16.59 years). All patients underwent complete physical examination, pulmonary computed tomography, laboratory tests, D-dimer, and blood gas analysis. There were no differences in laboratory tests or D-dimer. In patients with CPE, pO2, alveolar-arterial oxygen difference (A-aDO2), oxygen saturation %, and the ratio between arterial partial pressure of oxygen (PaO2) and fraction of inspired oxygen (FiO2), P/F, were significantly increased. There were no differences in PaCO2. Platelet count was inversely correlated to P/F (r = - 0.389, p = 0.02) but directly to A-aDO2 (r = 0.699, p = 0.001) only in patients with CPE. Histology of lung biopsies (7 CPE/7 controls) of patients with CPE showed an increase in CD15+ cells, HMGB1, and extracellular MPO as a marker of NETosis, while no significant differences were found in CD3+, CD4+, CD8+, and intracellular MPO. Overall, data suggest that CPE has a different clinical setting. Reduced oxygen content and saturation described in Patients with CPE should not be considered a trustworthy sign of disease. Increased A-aDO2 may indicate that CPE involves the smallest vessels as compared to classical PE. The significant difference in NETosis may suggest the mechanism related to thrombi formation.
Collapse
Affiliation(s)
- Sebastiano Cicco
- COVID Section, Unit of Internal Medicine "Guido Baccelli", Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), U.O.C. Medicina Interna "Guido Baccelli", University of Bari Aldo Moro, Policlinico di Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Antonio Vacca
- Division of Internal Medicine, Department of Medicine, University of Udine, Building 8, 33100, Udine (UD), Italy
| | - Federica Albanese
- COVID Section, Unit of Internal Medicine "Guido Baccelli", Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), U.O.C. Medicina Interna "Guido Baccelli", University of Bari Aldo Moro, Policlinico di Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Nicola Susca
- COVID Section, Unit of Internal Medicine "Guido Baccelli", Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), U.O.C. Medicina Interna "Guido Baccelli", University of Bari Aldo Moro, Policlinico di Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Vanessa Desantis
- COVID Section, Unit of Internal Medicine "Guido Baccelli", Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), U.O.C. Medicina Interna "Guido Baccelli", University of Bari Aldo Moro, Policlinico di Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
- Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), Pharmacology Section, University of Bari Aldo Moro Medical School, Bari, Italy
| | - Arianna Magistro
- COVID Section, Unit of Internal Medicine "Guido Baccelli", Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), U.O.C. Medicina Interna "Guido Baccelli", University of Bari Aldo Moro, Policlinico di Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Gerardo Cazzato
- Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), Section of Pathology, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Gerolamo Cicco
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41121, Modena, Italy
| | - Sara Sablone
- Department of Interdisciplinary Medicine, Section of Forensic Medicine, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - Christel Cariddi
- Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), Section of Anesthesiology and Intensive Care, Ospedale Policlinico, University of Bari Aldo Moro, Piazza G. Cesare 11, 70124, Bari, Italy
| | - Marialuisa Sveva Marozzi
- COVID Section, Unit of Internal Medicine "Guido Baccelli", Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), U.O.C. Medicina Interna "Guido Baccelli", University of Bari Aldo Moro, Policlinico di Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Cristiana Catena
- Division of Internal Medicine, Department of Medicine, University of Udine, Building 8, 33100, Udine (UD), Italy
| | - Gabriele Brosolo
- Division of Internal Medicine, Department of Medicine, University of Udine, Building 8, 33100, Udine (UD), Italy
| | - Stefano Marcante
- Division of Internal Medicine, Department of Medicine, University of Udine, Building 8, 33100, Udine (UD), Italy
| | - Giuseppe Ingravallo
- Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), Section of Pathology, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Lidia Dalfino
- Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), Section of Anesthesiology and Intensive Care, Ospedale Policlinico, University of Bari Aldo Moro, Piazza G. Cesare 11, 70124, Bari, Italy
| | - Gianfranco Lauletta
- COVID Section, Unit of Internal Medicine "Guido Baccelli", Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), U.O.C. Medicina Interna "Guido Baccelli", University of Bari Aldo Moro, Policlinico di Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Fabrizio Pappagallo
- COVID Section, Unit of Internal Medicine "Guido Baccelli", Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), U.O.C. Medicina Interna "Guido Baccelli", University of Bari Aldo Moro, Policlinico di Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Antonio Giovanni Solimando
- COVID Section, Unit of Internal Medicine "Guido Baccelli", Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), U.O.C. Medicina Interna "Guido Baccelli", University of Bari Aldo Moro, Policlinico di Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy.
| | - Salvatore Grasso
- Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), Section of Anesthesiology and Intensive Care, Ospedale Policlinico, University of Bari Aldo Moro, Piazza G. Cesare 11, 70124, Bari, Italy
| | - Eugenio Maiorano
- Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), Section of Pathology, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Francesco Introna
- Department of Interdisciplinary Medicine, Section of Forensic Medicine, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - Leonardo Alberto Sechi
- Division of Internal Medicine, Department of Medicine, University of Udine, Building 8, 33100, Udine (UD), Italy
| | - Roberto Ria
- COVID Section, Unit of Internal Medicine "Guido Baccelli", Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), U.O.C. Medicina Interna "Guido Baccelli", University of Bari Aldo Moro, Policlinico di Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| |
Collapse
|
249
|
Guo BC, Wu KH, Chen CY, Lin WY, Chang YJ, Lee TA, Lin MJ, Wu HP. Mesenchymal Stem Cells in the Treatment of COVID-19. Int J Mol Sci 2023; 24:14800. [PMID: 37834246 PMCID: PMC10573267 DOI: 10.3390/ijms241914800] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Since the emergence of the coronavirus disease 2019 (COVID-19) pandemic, many lives have been tragically lost to severe infections. The COVID-19 impact extends beyond the respiratory system, affecting various organs and functions. In severe cases, it can progress to acute respiratory distress syndrome (ARDS) and multi-organ failure, often fueled by an excessive immune response known as a cytokine storm. Mesenchymal stem cells (MSCs) have considerable potential because they can mitigate inflammation, modulate immune responses, and promote tissue regeneration. Accumulating evidence underscores the efficacy and safety of MSCs in treating severe COVID-19 and ARDS. Nonetheless, critical aspects, such as optimal routes of MSC administration, appropriate dosage, treatment intervals, management of extrapulmonary complications, and potential pediatric applications, warrant further exploration. These research avenues hold promise for enriching our understanding and refining the application of MSCs in confronting the multifaceted challenges posed by COVID-19.
Collapse
Affiliation(s)
- Bei-Cyuan Guo
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan;
| | - Kang-Hsi Wu
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 40201, Taiwan;
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Chun-Yu Chen
- Department of Emergency Medicine, Tungs’ Taichung Metro Harbor Hospital, Taichung 43503, Taiwan;
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 35664, Taiwan
| | - Wen-Ya Lin
- Department of Pediatrics, Taichung Veterans General Hospital, Taichung 43503, Taiwan
| | - Yu-Jun Chang
- Laboratory of Epidemiology and Biostastics, Changhua Christian Hospital, Changhua 50006, Taiwan;
| | - Tai-An Lee
- Department of Emergency Medicine, Chang Bing Show Chwan Memorial Hospital, Changhua 50544, Taiwan;
| | - Mao-Jen Lin
- Division of Cardiology, Department of Medicine, Taichung Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Taichung 42743, Taiwan
- Department of Medicine, College of Medicine, Tzu Chi University, Hualien 97002, Taiwan
| | - Han-Ping Wu
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Pediatrics, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| |
Collapse
|
250
|
Sayed AA, Al Nozha OM. Developing a COVID-19 Mortality Prediction (CoMPred) Indicator for ICU Diabetic Patients Treated with Tocilizumab in Saudi Arabia: A Proof-of-Concept Study. Biomedicines 2023; 11:2649. [PMID: 37893025 PMCID: PMC10603829 DOI: 10.3390/biomedicines11102649] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Since the beginning of the COVID-19 pandemic, efforts have been made to underline its discourse and identify factors contributing to its severe forms. Clinically, many physicians depended on subjective criteria to determine its severe forms, which varied significantly between practices. However, they did not rely on objective laboratory findings. This study aimed to present a novel and objective laboratory-based indicator to predict mortality among COVID-19 patients. The study included 249 COVID-19 patients who were admitted to the ICU, of which 80 did not survive. The COVID-19 Mortality Prediction (CoMPred) indicator was developed by including the age and the following lab investigations: neutrophil-to-lymphocyte ratio (NLR), D-Dimer, PT, aPTT, ESR, CRP, and urea levels. A CoMPred score of 7.5 or higher carries a sensitivity of 81.10% in predicting mortality, i.e., a patient with a CoMPred score of 7.5 or higher has an 81.10% chance of dying. The CoMPred indicator score directly correlates with mortality, i.e., the higher the score, the higher the possibility of the patient dying. In conclusion, the CoMPred indicator is an objective tool that is affordable and widely available, will assist physicians, and limit the burden on clinical decisions on an unpredicted course of COVID-19 in patients.
Collapse
Affiliation(s)
- Anwar A. Sayed
- Department of Medical Microbiology and Immunology, College of Medicine, Taibah University, Madina 42353, Saudi Arabia
| | - Omar M. Al Nozha
- Department of Medicine, Taibah University, Madina 42353, Saudi Arabia
- Department of Medicine, Saudi German Hospital, Madina 42373, Saudi Arabia
| |
Collapse
|