51
|
Atmaca MM, Bülbül NG, Ateş MF, Selbest B, Eren F, Güler S, Şimşek UB, Yetkin MF, Akcakoyunlu M, Biçer EÖ, Kılıç S, Yetkin NA, Taş Ş, Kocakaya Z, Karasu K, Yüksel U, Kılıç AU, Yüce ZT, Tutar N, Ketencioğlu BB, Gündoğan K, Temel Ş, Hasan MA, Benli Ş, Eröz Ç, Öztürk A, Erdoğan FF. Incidence and Features of Acute Ischemic Stroke in Patients Hospitalized with COVID-19: A Multi-center Study in Turkey. Noro Psikiyatr Ars 2024; 67:241-247. [PMID: 39258124 PMCID: PMC11382562 DOI: 10.29399/npa.28493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/03/2023] [Indexed: 09/12/2024] Open
Abstract
Introduction Acute ischemic stroke (AIS) is a devastating complication of COVID-19 with high morbidity and mortality. In this study, we reported the frequency, characteristics, and outcome of AIS in patients with COVID-19. Methods This multicenter and cross-sectional study was conducted between April 2020 and February 2021. Among the hospitalized patients with COVID-19, the detailed characteristics of those with and without AIS were recorded and compared. Results Six hundred ninety-three patients were included in the study. Acute ischemic stroke was detected in 16 (2.31%) patients, the median age was 77 (range, 48-91) years, and 10 (62.5%) were female. The median NIHSS score at admission was 9 (range, 3-17). Total anterior circulation infarction (TACI) was the most common (37.5%) type and cardioembolism was the most common etiology (37.5%). Nine patients (56.25%) developed AIS within 24 hours of having COVID-19. COVID-19 severity was severe or critical in seven patients (43.75%). Eight patients died, and eight were discharged. Patients with AIS had a higher rate of hypertension, coronary artery disease, heart failure, a history of myocardial infarction, a history of cerebrovascular disease, severe and critical COVID-19, a higher mean age, and a longer ICU stay compared with those without AIS (p<0.001 for each). Conclusions AIS can occur in patients with COVID-19 and is associated with mortality. Acute ischemic stroke is encountered at any stage of COVID-19, especially within the first 72 hours of the diagnosis, in older patients with comorbidities and severe COVID-19. There is an increased risk of AIS in patients with COVID-19 with a history of stroke.
Collapse
Affiliation(s)
- Murat Mert Atmaca
- Sultan 2.Abdulhamid Khan Training and Research Hospital Neurology Clinic, Turkey
| | - Nazlı Gamze Bülbül
- Sultan 2.Abdulhamid Khan Training and Research Hospital Neurology Clinic, Turkey
| | | | | | - Fettah Eren
- Selçuk University, Faculty of Medicine, Department of Neurology, Turkey
| | - Sibel Güler
- Trakya University, Faculty of Medicine, Department of Neurology, Turkey
| | - Uğur Burak Şimşek
- Erciyes University, Faculty of Medicine, Department of Neurology, Turkey
| | | | - Merve Akcakoyunlu
- Erciyes University, Faculty of Medicine, Department of Neurology, Turkey
| | - Elif Özge Biçer
- Erciyes University, Faculty of Medicine, Department of Neurology, Turkey
| | - Soner Kılıç
- Kayseri City Hospital Neurology Clinic, Turkey
| | | | - Şazimet Taş
- Erciyes University, Faculty of Medicine, Department of Internal Medicine, Intensive Care Unit, Turkey
| | - Zeynep Kocakaya
- Erciyes University, Faculty of Medicine, Department of Internal Medicine, Intensive Care Unit, Turkey
| | - Kevser Karasu
- Erciyes University, Faculty of Medicine, Department of Internal Medicine, Intensive Care Unit, Turkey
| | - Ufuk Yüksel
- Erciyes University, Faculty of Medicine, Department of Internal Medicine, Intensive Care Unit, Turkey
| | - Ayşegül Ulu Kılıç
- Erciyes University, Faculty of Medicine, Department of Infectious Diseases, Turkey
| | - Zeynep Türe Yüce
- Erciyes University, Faculty of Medicine, Department of Infectious Diseases, Turkey
| | - Nuri Tutar
- Erciyes University, Faculty of Medicine Department of Chest Diseases, Turkey
| | | | - Kürşat Gündoğan
- Erciyes University, Faculty of Medicine, Department of Internal Medicine, Intensive Care Unit, Turkey
| | - Şahin Temel
- Erciyes University, Faculty of Medicine, Department of Internal Medicine, Intensive Care Unit, Turkey
| | - Murad Al Hasan
- Erciyes University, Faculty of Medicine, Department of Neurology, Turkey
| | - Şeyma Benli
- Erciyes University, Faculty of Medicine, Department of Neurology, Turkey
| | - Çağla Eröz
- Erciyes University, Faculty of Medicine, Department of Neurology, Turkey
| | - Ahmet Öztürk
- Erciyes University, Faculty of Medicine, Department of Biostatistics, Turkey
| | | |
Collapse
|
52
|
Satoh K, Wada T, Tampo A, Takahashi G, Hoshino K, Matsumoto H, Taira T, Kazuma S, Masuda T, Tagami T, Ishikura H. Practical approach to thrombocytopenia in patients with sepsis: a narrative review. Thromb J 2024; 22:67. [PMID: 39039520 PMCID: PMC11265094 DOI: 10.1186/s12959-024-00637-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
Thrombocytopenia frequently occurs in patients with sepsis. Disseminated intravascular coagulation (DIC) may be a possible cause of thrombocytopenia owing to its high prevalence and association with poor outcomes; however, it is important to keep the presence of other diseases in mind in sepsis practice. Thrombotic microangiopathy (TMA), which is characterized by thrombotic thrombocytopenic purpura, Shiga toxin-producing Escherichia coli hemolytic uremic syndrome (HUS), and complement-mediated HUS, is characterized by thrombocytopenia, microangiopathic hemolytic anemia, and organ damage. TMA has become widely recognized in recent years because of the development of specific treatments. Previous studies have reported a remarkably lower prevalence of TMA than DIC; however, its epidemiology is not well defined, and there may be cases in which TMA is not correctly diagnosed, resulting in poor outcomes. Therefore, it is important to differentiate DIC from TMA. Nevertheless, differentiating between DIC and TMA remains a challenge as indicated by previous reports that most patients with TMA can be diagnosed as DIC using the universal coagulation scoring system. Several algorithms to differentiate sepsis-related DIC from TMA have been suggested, contributing to improving the care of septic patients with thrombocytopenia; however, it may be difficult to apply these algorithms to patients with coexisting DIC and TMA, which has recently been reported. This review describes the disease characteristics, including epidemiology, pathophysiology, and treatment, of DIC, TMA, and other diseases with thrombocytopenia and proposes a novel practical approach flow, which is characterized by the initiation of the diagnosis of TMA in parallel with the diagnosis of DIC. This practical flow also refers to the longitudinal diagnosis and treatment flow with TMA in mind and real clinical timeframes. In conclusion, we aim to widely disseminate the results of this review that emphasize the importance of incorporating consideration of TMA in the management of septic DIC. We anticipate that this practical new approach for the diagnostic and treatment flow will lead to the appropriate diagnosis and treatment of complex cases, improve patient outcomes, and generate new epidemiological evidence regarding TMA.
Collapse
Affiliation(s)
- Kasumi Satoh
- Advanced Emergency and Critical Care Center, Akita University Hospital, Akita, Japan
| | - Takeshi Wada
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Faculty of Medicine, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Akihito Tampo
- Department of Emergency Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Gaku Takahashi
- Department of Critical Care, Disaster and General Medicine, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Kota Hoshino
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Hironori Matsumoto
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Toon, Japan
| | - Takayuki Taira
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Satoshi Kazuma
- Department of Intensive Care Medicine, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Takamitsu Masuda
- Department of Emergency Medicine, Emergency and Critical Care Center, Fujieda Municipal General Hospital, Fujieda, Japan
| | - Takashi Tagami
- Department of Emergency and Critical Care Medicine, Nippon Medical School Musashikosugi Hospital, Tokyo, Japan
| | - Hiroyasu Ishikura
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
53
|
Wang H, Wu S, Pan D, Meng W, Hu L, Zhang H, Ning Y, Guo J, Gu Y. Causal relationships between COVID-19 and venous thromboembolism: A mendelian randomization analysis. Phlebology 2024:2683555241266659. [PMID: 39033375 DOI: 10.1177/02683555241266659] [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: 07/23/2024]
Abstract
Objective: Observational studies show the correlation between COVID-19 and venous thromboembolism (VTE) risk. However, the causal effects remain uncertain. We aimed to explore the potential causal association between COVID-19 and VTE using Mendelian randomization (MR) design. Methods: Two-sample MR was used to evaluate the potential causality between COVID-19 and VTE by selecting single-nucleotide polymorphisms (SNPs) as instrumental variables (IVs) from genome-wide association studies (GWAS). The weighted median, MR-Egger, simple mode, and weighted mode were employed as supplementary methods for MR estimations, with the inverse-variance weighted (IVW) method serving as the principal analysis. In addition, we took sensitivity analyses, including Cochran's test, MR-Pleiotropy Residual Sum and Outlier (MR-PRESSO), and leave-one-out analysis to ensure that we obtained stable and reliable results. Results: Our study selected 26 COVID-19 severity, 31 COVID-19 hospitalization, and 13 COVID-19 susceptibility SNPs as instrumental variables. The IVW analysis results revealed that there was no causal relationship between COVID-19 severity, hospitalization, or susceptibility and VTE, with odds ratios of 0.974 (95%CI: 0.936-1.013, p = 0.19), 0.976 (95%CI: 0.918-1.039, p = 0.447), and 0.908 (95%CI: 0.775-1.065, p = 0.235), respectively. The IVW approach yielded consistent results with MR-Egger, Weighted Median simple mode, and weighted mode. MR-PRESSO and sensitivity analysis further confirmed the stability and consistency of the MR results. Conclusions: This study did not find evidence to support a causal relationship between COVID-19 and VTE at the genetic level. Further investigation is warranted to determine if the significant association reported in previous observational studies between the two is due to confounding factors.
Collapse
Affiliation(s)
- Hui Wang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Sensen Wu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dikang Pan
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenzhuo Meng
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Lefan Hu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hanyu Zhang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yachan Ning
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jianming Guo
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yongquan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
54
|
Shukla N, Shamim U, Agarwal P, Pandey R, Narayan J. From bench to bedside: potential of translational research in COVID-19 and beyond. Brief Funct Genomics 2024; 23:349-362. [PMID: 37986554 DOI: 10.1093/bfgp/elad051] [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: 09/07/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease 2019 (COVID-19) have been around for more than 3 years now. However, due to constant viral evolution, novel variants are emerging, leaving old treatment protocols redundant. As treatment options dwindle, infection rates continue to rise and seasonal infection surges become progressively common across the world, rapid solutions are required. With genomic and proteomic methods generating enormous amounts of data to expand our understanding of SARS-CoV-2 biology, there is an urgent requirement for the development of novel therapeutic methods that can allow translational research to flourish. In this review, we highlight the current state of COVID-19 in the world and the effects of post-infection sequelae. We present the contribution of translational research in COVID-19, with various current and novel therapeutic approaches, including antivirals, monoclonal antibodies and vaccines, as well as alternate treatment methods such as immunomodulators, currently being studied and reiterate the importance of translational research in the development of various strategies to contain COVID-19.
Collapse
Affiliation(s)
- Nityendra Shukla
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Near Jubilee Hall, New Delhi, 110007, India
| | - Uzma Shamim
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Near Jubilee Hall, New Delhi, 110007, India
| | - Preeti Agarwal
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Near Jubilee Hall, New Delhi, 110007, India
| | - Rajesh Pandey
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Near Jubilee Hall, New Delhi, 110007, India
| | - Jitendra Narayan
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Near Jubilee Hall, New Delhi, 110007, India
| |
Collapse
|
55
|
Kim HJ, Jeong S, Song J, Park SJ, Park YJ, Oh YH, Jung J, Park SM. Risk of pulmonary embolism and deep vein thrombosis following COVID-19: a nationwide cohort study. MedComm (Beijing) 2024; 5:e655. [PMID: 39006761 PMCID: PMC11246596 DOI: 10.1002/mco2.655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 07/16/2024] Open
Abstract
Recent studies elucidate that coronavirus disease 2019 (COVID-19) patients may face a higher risk of cardiovascular complications. This study aimed to evaluate association of COVID-19 with the risk of pulmonary embolism (PE) or deep vein thrombosis (DVT). This nationwide population-based retrospective cohort study included Korean adult citizens between January 2021 and March 2022 from the Korea Disease Control and Prevention Agency COVID-19 National Health Insurance Service cohort. The Fine and Gray's regression with all-cause death as a competing event was adopted to evaluate PE and DVT risks after COVID-19. This study included a total of 1,601,835 COVID-19 patients and 14,011,285 matched individuals without COVID-19. The risk of PE (adjusted hazard ratio [aHR], 6.25; 95% confidence interval [CI], 3.67-10.66; p < 0.001) and DVT (aHR, 3.05; 95% CI, 1.75-5.29; p < 0.001) was higher in COVID-19 group in individuals without complete COVID-19 vaccination. In addition, individuals with complete COVID-19 vaccination still had a higher risk of COVID-19-related PE (aHR, 1.48; 95% CI, 1.15-1.88; p < 0.001). However, COVID-19 was not a significant risk factor for DVT among those with complete COVID-19 vaccination. COVID-19 was identified as an independent factor that elevated PE and DVT risks, especially for individuals without complete COVID-19 vaccination.
Collapse
Affiliation(s)
- Hye Jun Kim
- Department of Biomedical Sciences Seoul National University College of Medicine Seoul South Korea
| | - Seogsong Jeong
- Department of Biomedical Informatics Korea University College of Medicine Seoul South Korea
| | - Jihun Song
- Department of Biomedical Sciences Seoul National University College of Medicine Seoul South Korea
| | - Sun Jae Park
- Department of Biomedical Sciences Seoul National University College of Medicine Seoul South Korea
| | - Young Jun Park
- Medical Research Center Genomic Medicine Institute Seoul National University Seoul South Korea
| | - Yun Hwan Oh
- Department of Family Medicine Chung-Ang University Gwangmyeong Hospital Chung-Ang University College of Medicine Gwangmyeong South Korea
| | - Jaehun Jung
- Department of Preventive Medicine Gachon University College of Medicine Incheon South Korea
| | - Sang Min Park
- Department of Biomedical Sciences Seoul National University College of Medicine Seoul South Korea
- Department of Family Medicine Seoul National University Hospital Seoul South Korea
| |
Collapse
|
56
|
Kouchek M, Miri MM, Aghakhani K, Memarian A. ABO blood group is related to the prevalence, severity, and mortality rate of COVID-19. MEDICINA CLÍNICA PRÁCTICA 2024; 7:100442. [DOI: 10.1016/j.mcpsp.2024.100442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
57
|
Pacheco G, Lopes ALF, Oliveira APD, Corrêa WDRM, Lima LDB, Souza MHLPD, Teles AS, Nicolau LAD, Medeiros JVR. Comprehensive analysis of gastrointestinal side effects in COVID-19 patients undergoing combined pharmacological treatment with azithromycin and hydroxychloroquine: a systematic review and network meta-analysis. Crit Rev Toxicol 2024; 54:345-358. [PMID: 38860720 DOI: 10.1080/10408444.2024.2348169] [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: 01/29/2024] [Accepted: 04/22/2024] [Indexed: 06/12/2024]
Abstract
During the COVID-19 pandemic, several drugs were repositioned and combined to quickly find a way to mitigate the effects of the infection. However, the adverse effects of these combinations on the gastrointestinal tract are unknown. We aimed investigate whether Hydroxychloroquine (HD), Azithromycin (AZ), and Ivermectin (IV) used in combination for the treatment of COVID-19, can lead to the development of gastrointestinal disorders. This is a systematic review and network meta-analysis conducted using Stata and Revman software, respectively. The protocol was registered with PROSPERO (CRD42023372802). A search of clinical trials in Cochrane Library databases, Embase, Web of Science, Lilacs, PubMed, Scopus and Clinicaltrials.gov conducted on November 26, 2023. The eligibility of the studies was assessed based on PICO criteria, including trials that compared different treatments and control group. The analysis of the quality of the evidence was carried out according to the GRADE. Six trials involving 1,686 COVID-19 patients were included. No trials on the association of HD or AZ with IV met the inclusion criteria, only studies on the association between HD and AZ were included. Nausea, vomiting, diarrhea, abdominal pain and increased transaminases were related. The symptoms of vomiting and nausea were evaluated through a network meta-analysis, while the symptom of abdominal pain was evaluated through a meta-analysis. No significant associations with these symptoms were observed for HD, AZ, or their combination, compared to control. Low heterogeneity and absence of inconsistency in indirect and direct comparisons were noted. Limitations included small sample sizes, varied drug dosages, and potential publication bias during the pandemic peak. This review unveils that there are no associations between gastrointestinal adverse effects and the combined treatment of HD with AZ in the management of COVID-19, as compared to either the use of a control group or the administration of the drugs individually, on the other hand, highlighting the very low or low certainty of evidence for the evaluated outcomes. To accurately conclude the absence of side effects, further high-quality randomized studies are needed.
Collapse
Affiliation(s)
- Gabriella Pacheco
- Medicinal Plants Research Center (NPPM), Post-graduation Program in Pharmacology, Federal University of Piauí, Teresina, PI, Brazil
| | - André Luis Fernandes Lopes
- Biotechnology and Biodiversity Research Center (BIOTEC), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University (UFDPar), Parnaíba, PI, Brazil
| | | | | | - Lucas Daniel Batista Lima
- Biotechnology and Biodiversity Research Center (BIOTEC), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University (UFDPar), Parnaíba, PI, Brazil
| | | | - Ariel Soares Teles
- Biotechnology and Biodiversity Research Center (BIOTEC), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University (UFDPar), Parnaíba, PI, Brazil
- Federal Institute of Maranhão (IFMA), Araioses, MA, Brazil
| | - Lucas Antonio Duarte Nicolau
- Biotechnology and Biodiversity Research Center (BIOTEC), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University (UFDPar), Parnaíba, PI, Brazil
| | - Jand Venes Rolim Medeiros
- Medicinal Plants Research Center (NPPM), Post-graduation Program in Pharmacology, Federal University of Piauí, Teresina, PI, Brazil
- Biotechnology and Biodiversity Research Center (BIOTEC), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University (UFDPar), Parnaíba, PI, Brazil
| |
Collapse
|
58
|
Zhao R, Li M, Xiao P, Song D, Li H. Advances in D-dimer testing: progress in harmonization of clinical assays and innovative detection methods. Anal Bioanal Chem 2024; 416:3737-3750. [PMID: 38503987 DOI: 10.1007/s00216-024-05207-x] [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: 12/08/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 03/21/2024]
Abstract
The D-dimer is a sensitive indicator of coagulation and fibrinolysis activation, especially valuable as a biomarker of intravascular thrombosis. Measurement of plasma D-dimer levels plays a crucial role in the diagnosis and monitoring of conditions such as deep vein thrombosis, pulmonary embolism, and disseminated intravascular coagulation. A variety of immunoassays, including enzyme-linked immunosorbent assays, latex-enhanced immunoturbidimetric assays, whole-blood aggregation analysis, and immunochromatography assays, are widely used in clinical settings to determine D-dimer levels. However, the results obtained from different D-dimer assays vary significantly. These assays exhibit intra-method coefficients of variation ranging from 6.4% to 17.7%, and the measurement discrepancies among different assays can be as high as 20-fold. The accuracy and reliability of D-dimer testing cannot be guaranteed due to the lack of an internationally endorsed reference measurement system (including reference materials and reference measurement procedures), which may lead to misdiagnosis and underdiagnosis, limiting its full clinical application. In this review, we present an in-depth analysis of clinical D-dimer testing, summarizing the existing challenges, the current state of metrology, and progress towards harmonization. We also review the latest advancements in D-dimer detection techniques, which include mass spectrometry and electrochemical and optical immunoassays. By comparing the basic principles, the definition of the measurand, and analytical performance of these methods, we provide an outlook on the potential improvements in D-dimer clinical testing.
Collapse
Affiliation(s)
- Rong Zhao
- National Institute of Metrology, Beijing, 100029, China
- Key Laboratory of Chemical Metrology and Applications on Nutrition and Health for State Market Regulation, Beijing, 100029, China
| | - Mengran Li
- Beijing University of Chemical Technology, Beijing, 100029, China
| | - Peng Xiao
- National Institute of Metrology, Beijing, 100029, China
- Key Laboratory of Chemical Metrology and Applications on Nutrition and Health for State Market Regulation, Beijing, 100029, China
| | - Dewei Song
- National Institute of Metrology, Beijing, 100029, China
| | - Hongmei Li
- National Institute of Metrology, Beijing, 100029, China.
- Key Laboratory of Chemical Metrology and Applications on Nutrition and Health for State Market Regulation, Beijing, 100029, China.
| |
Collapse
|
59
|
Li X, Mi Z, Liu Z, Rong P. SARS-CoV-2: pathogenesis, therapeutics, variants, and vaccines. Front Microbiol 2024; 15:1334152. [PMID: 38939189 PMCID: PMC11208693 DOI: 10.3389/fmicb.2024.1334152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/29/2024] [Indexed: 06/29/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged in December 2019 with staggering economic fallout and human suffering. The unique structure of SARS-CoV-2 and its underlying pathogenic mechanism were responsible for the global pandemic. In addition to the direct damage caused by the virus, SARS-CoV-2 triggers an abnormal immune response leading to a cytokine storm, culminating in acute respiratory distress syndrome and other fatal diseases that pose a significant challenge to clinicians. Therefore, potential treatments should focus not only on eliminating the virus but also on alleviating or controlling acute immune/inflammatory responses. Current management strategies for COVID-19 include preventative measures and supportive care, while the role of the host immune/inflammatory response in disease progression has largely been overlooked. Understanding the interaction between SARS-CoV-2 and its receptors, as well as the underlying pathogenesis, has proven to be helpful for disease prevention, early recognition of disease progression, vaccine development, and interventions aimed at reducing immunopathology have been shown to reduce adverse clinical outcomes and improve prognosis. Moreover, several key mutations in the SARS-CoV-2 genome sequence result in an enhanced binding affinity to the host cell receptor, or produce immune escape, leading to either increased virus transmissibility or virulence of variants that carry these mutations. This review characterizes the structural features of SARS-CoV-2, its variants, and their interaction with the immune system, emphasizing the role of dysfunctional immune responses and cytokine storm in disease progression. Additionally, potential therapeutic options are reviewed, providing critical insights into disease management, exploring effective approaches to deal with the public health crises caused by SARS-CoV-2.
Collapse
Affiliation(s)
- Xi Li
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ze Mi
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhenguo Liu
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
60
|
Lu W, Yan L, Tang X, Wang X, Du J, Zou Z, Li L, Ye J, Zhou L. Efficacy and safety of mesenchymal stem cells therapy in COVID-19 patients: a systematic review and meta-analysis of randomized controlled trials. J Transl Med 2024; 22:550. [PMID: 38851730 PMCID: PMC11162060 DOI: 10.1186/s12967-024-05358-6] [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: 04/14/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) has become a serious public health issue. In COVID-19 patients, the elevated levels of inflammatory cytokines lead to the manifestation of COVID-19 symptoms, such as lung tissue edema, lung diffusion dysfunction, acute respiratory distress syndrome (ARDS), secondary infection, and ultimately mortality. Mesenchymal stem cells (MSCs) exhibit anti-inflammatory and immunomodulatory properties, thus providing a potential treatment option for COVID-19. The number of clinical trials of MSCs for COVID-19 has been rising. However, the treatment protocols and therapeutic effects of MSCs for COVID-19 patients are inconsistent. This meta-analysis was performed to systematically determine the safety and efficacy of MSC infusion in COVID-19 patients. METHODS We conducted a comprehensive literature search from PubMed/Medline, Web of Science, EMBASE, and Cochrane Library up to 22 November 2023 to screen for eligible randomized controlled trials. Inclusion and exclusion criteria for searched literature were formulated according to the PICOS principle, followed by the use of literature quality assessment tools to assess the risk of bias. Finally, outcome measurements including therapeutic efficacy, clinical symptoms, and adverse events of each study were extracted for statistical analysis. RESULTS A total of 14 randomized controlled trials were collected. The results of enrolled studies demonstrated that patients with COVID-19 pneumonia who received MSC inoculation showed a decreased mortality compared with counterparts who received conventional treatment (RR: 0.76; 95% CI [0.60, 0.96]; p = 0.02). Reciprocally, MSC inoculation improved the clinical symptoms in patients (RR: 1.28; 95% CI [1.06, 1.55]; p = 0.009). In terms of immune biomarkers, MSC treatment inhibited inflammation responses in COVID-19 patients, as was indicated by the decreased levels of CRP and IL-6. Importantly, our results showed that no significant differences in the incidence of adverse reactions or serious adverse events were monitored in patients after MSC inoculation. CONCLUSION This meta-analysis demonstrated that MSC inoculation is effective and safe in the treatment of patients with COVID-19 pneumonia. Without increasing the incidence of adverse events or serious adverse events, MSC treatment decreased patient mortality and inflammatory levels and improved the clinical symptoms in COVID-19 patients. However, large-cohort randomized controlled trials with expanded numbers of patients are required to further confirm our results.
Collapse
Affiliation(s)
- Wenming Lu
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- School of Rehabilitation Medicine, Gannan Medical University, GanZhou City, 341000, Jiangxi, People's Republic of China
- The First Clinical College of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Longxiang Yan
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- School of Rehabilitation Medicine, Gannan Medical University, GanZhou City, 341000, Jiangxi, People's Republic of China
- The First Clinical College of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Xingkun Tang
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- School of Rehabilitation Medicine, Gannan Medical University, GanZhou City, 341000, Jiangxi, People's Republic of China
| | - Xuesong Wang
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- School of Rehabilitation Medicine, Gannan Medical University, GanZhou City, 341000, Jiangxi, People's Republic of China
| | - Jing Du
- School of Rehabilitation Medicine, Gannan Medical University, GanZhou City, 341000, Jiangxi, People's Republic of China
| | - Zhengwei Zou
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- Ganzhou Key Laboratory of Stem Cell and Regenerative Medicine, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Lincai Li
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- Ganzhou Key Laboratory of Stem Cell and Regenerative Medicine, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Junsong Ye
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- Ganzhou Key Laboratory of Stem Cell and Regenerative Medicine, Ganzhou, 341000, Jiangxi, People's Republic of China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- Jiangxi Provincal Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Lin Zhou
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China.
- Ganzhou Key Laboratory of Stem Cell and Regenerative Medicine, Ganzhou, 341000, Jiangxi, People's Republic of China.
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China.
- Jiangxi Provincal Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China.
| |
Collapse
|
61
|
Carvajal JJ, García-Castillo V, Cuellar SV, Campillay-Véliz CP, Salazar-Ardiles C, Avellaneda AM, Muñoz CA, Retamal-Díaz A, Bueno SM, González PA, Kalergis AM, Lay MK. New insights into the pathogenesis of SARS-CoV-2 during and after the COVID-19 pandemic. Front Immunol 2024; 15:1363572. [PMID: 38911850 PMCID: PMC11190347 DOI: 10.3389/fimmu.2024.1363572] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 04/24/2024] [Indexed: 06/25/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the respiratory distress condition known as COVID-19. This disease broadly affects several physiological systems, including the gastrointestinal, renal, and central nervous (CNS) systems, significantly influencing the patient's overall quality of life. Additionally, numerous risk factors have been suggested, including gender, body weight, age, metabolic status, renal health, preexisting cardiomyopathies, and inflammatory conditions. Despite advances in understanding the genome and pathophysiological ramifications of COVID-19, its precise origins remain elusive. SARS-CoV-2 interacts with a receptor-binding domain within angiotensin-converting enzyme 2 (ACE2). This receptor is expressed in various organs of different species, including humans, with different abundance. Although COVID-19 has multiorgan manifestations, the main pathologies occur in the lung, including pulmonary fibrosis, respiratory failure, pulmonary embolism, and secondary bacterial pneumonia. In the post-COVID-19 period, different sequelae may occur, which may have various causes, including the direct action of the virus, alteration of the immune response, and metabolic alterations during infection, among others. Recognizing the serious adverse health effects associated with COVID-19, it becomes imperative to comprehensively elucidate and discuss the existing evidence surrounding this viral infection, including those related to the pathophysiological effects of the disease and the subsequent consequences. This review aims to contribute to a comprehensive understanding of the impact of COVID-19 and its long-term effects on human health.
Collapse
Affiliation(s)
- Jonatan J. Carvajal
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | - Valeria García-Castillo
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | - Shelsy V. Cuellar
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | | | - Camila Salazar-Ardiles
- Center for Research in Physiology and Altitude Medicine (FIMEDALT), Biomedical Department, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Andrea M. Avellaneda
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Department of Basic Sciences, Faculty of Sciences, Universidad Santo Tomás, Antofagasta, Chile
| | - Christian A. Muñoz
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Angello Retamal-Díaz
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K. Lay
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| |
Collapse
|
62
|
Ertan-Bolelli T, Bolelli K, Elçi SD, Belen-Apak FB. Promising Drug Fondaparinux for the Treatment of COVID-19: an In Silico Analysis of Low Molecular Weight Heparin, Direct Oral Anticoagulant, and Antiplatelet Drug Interactions with Host Protease Furin. Cardiovasc Drugs Ther 2024; 38:425-432. [PMID: 36401727 PMCID: PMC9676724 DOI: 10.1007/s10557-022-07406-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/07/2022] [Indexed: 11/21/2022]
Abstract
PURPOSE As of July 2022, the COVID-19 pandemic has affected over 555 million worldwide confirmed cases and caused more than 6.3 million deaths. The studies showed that the D-dimer levels were increased in non-survivors compared to survivors and heparin treatment has begun to be administered to the patients in severe clinics. As we knew that the entrance of SARS-CoV-2 to the host cell needs to be facilitated by host proteases; we published our hypothesis that heparin as a serine protease inhibitor may block the interaction between spike protein receptor-binding domain and host proteases. In our study, we aimed to investigate the interactions between not only heparins but also other antiplatelet and anticoagulant drugs including fondaparinux. METHODS In this study, docking studies were carried out to evaluate the interactions between low molecular weight heparins (LMWHs) (enoxaparin, dalteparin, tinzaparin), direct oral anticoagulant, and antiplatelet drugs with host proteases. Molecular docking studies were performed by using Schrödinger molecular modeling software. 3D structures of the ligands were obtained from the 2D structures by assigning the OPLS-2005 force field using the Maestro 12.7. The 3D crystal structure of the furin complexed with an inhibitor, 2,5-dideoksistreptamin derivative, was extracted from the Protein Data Bank (PDB ID: 5MIM). Docking studies were carried out using the Grid-based Ligand Docking with Energetics module of the Schrödinger Software. RESULTS The docking studies revealed that fondaparinux was the most relevant molecule to interact with furin with a docking score of - 12.74. It showed better interaction than the natural ligand of furin with an increased score compared to the docking score of - 8.155 of the natural ligand. AnaGA*IsA structure representing LMWH structure has shown a docking score of - 11.562 which was also better than the score of the natural ligand of furin. CONCLUSION Our findings have shown that LMWHs and fondaparinux can be used for their possible antiviral effects in COVID-19 patients. Our results have shown that in accordance with heparin and LMWH, fondaparinux can also be a candidate for "drug repurposing" in COVID-19 therapy, not only because of their anticoagulant but also possible antiviral effects.
Collapse
Affiliation(s)
- Tugba Ertan-Bolelli
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey
- Bolelli Lab LLC, Stone Mountain, GA 30083 USA
| | - Kayhan Bolelli
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey
- Bolelli Lab LLC, Stone Mountain, GA 30083 USA
- LumiLabs, Ulus, Ankara, 06610 Turkey
| | | | - F. Burcu Belen-Apak
- Department of Pediatric Hematology and Oncology, Medical Faculty, Baskent University, Sehit Temel Kuguluoglu Street No 24, 06490 Bahcelievler/Ankara, Turkey
| |
Collapse
|
63
|
Mathew S, Ramaswamy S, P V S, Kalanad A, John AG. Retrospective Analysis of Acute Kidney Injury in COVID-19 Infection: A Single-Center Study From Kerala. Cureus 2024; 16:e61772. [PMID: 38975470 PMCID: PMC11227431 DOI: 10.7759/cureus.61772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2024] [Indexed: 07/09/2024] Open
Abstract
Introduction The COVID-19 pandemic, caused by SARS-CoV-2, has resulted in significant morbidity and mortality worldwide. While primarily a respiratory illness, COVID-19 can lead to multi-organ involvement, including acute kidney injury (AKI). This study aimed to retrospectively analyze the incidence, risk factors, and outcomes of AKI in COVID-19 patients. Methods A single-center retrospective study involving 232 severe COVID-19 patients requiring ICU admission was analyzed. Patients were categorized into two groups based on renal involvement: group A (with AKI or worsening of pre-existing chronic kidney disease) and group B (without renal injury). Data on demographics, comorbidities, clinical presentation, inflammatory markers, management strategies, and outcomes were collected and analyzed. Results AKI or worsening of pre-existing chronic renal disease was noted in 50.87% of cases, while the remaining 49.13% had severe COVID-19 pneumonia without renal injury. The mean age of patients in group A (with renal involvement) was higher compared to group B (without renal injury), with a significant male predominance observed in group A. AKI occurred within a short duration of fever, and cough was not a significant symptom. Comorbidities such as diabetes, hypertension, and chronic kidney disease were common in both groups, with hypertension significantly associated with AKI. Other significant comorbidities as risk factors for kidney injury included chronic liver disease, coronary artery disease, chronic kidney disease, and malignancy. Elevated inflammatory markers, including C-reactive protein (CRP), serum ferritin, and interleukin-6, were significantly associated with renal injury. There was no significant difference in the mortality rate between the two groups studied. Conclusion AKI or worsening of pre-existing kidney disease is a common event in severe COVID-19 infection. Patients, especially elderly males with comorbidities as mentioned, should be thoroughly monitored for worsening renal function, and steps like avoidance of nephrotoxic drugs and timely hemodynamic support may help avoid this dreaded complication to a certain extent and improve the prognosis in severe COVID-19 infection. Supportive care remains crucial in managing COVID-19 patients with renal involvement, emphasizing the need for the early detection and treatment of renal abnormalities. Long-term follow-up is essential to assess the impact of AKI on future kidney health.
Collapse
Affiliation(s)
- Sheela Mathew
- Infectious Diseases, Government Medical College, Kozhikode, Kozhikode, IND
| | - Sreejith Ramaswamy
- Internal Medicine, Government Medical College, Kozhikode, Kozhikode, IND
| | - Shiji P V
- Internal Medicine, Government Medical College, Kozhikode, Kozhikode, IND
| | - Aquil Kalanad
- Internal Medicine, Government Medical College, Kozhikode, Kozhikode, IND
| | - Aaron G John
- Gastrointestinal, Hepatopancreatobiliary, and Multi-Organ Transplant Surgery, Rajagiri Hospital, Aluva, IND
| |
Collapse
|
64
|
Çakmak R, Yüce S, Ay M, Uyar MH, Kılıç Mİ, Bektaş M. Intravenous high-dose anakinra drops venous thrombosis and acute coronary syndrome in severe and critical COVID-19 patients: a propensity score matched study. Sci Rep 2024; 14:12369. [PMID: 38811592 PMCID: PMC11137068 DOI: 10.1038/s41598-024-62079-y] [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: 02/27/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024] Open
Abstract
In our study, we aimed to evaluate the effect of high-dose intravenous anakinra treatment on the development of thrombotic events in severe and critical COVID-19 patients. This retrospective observational study was conducted at a tertiary referral center in Aksaray, Turkey. The study population consisted of two groups as follows; the patients receiving high-dose intravenous anakinra (anakinra group) added to background therapy and the patients treated with standard of care (SoC) as a historical control group. Age, gender, mcHIS scores, and comorbidities such as diabetes mellitus, hypertension, and coronary heart disease of the patients were determined as the variables to be matched. We included 114 patients in SoC and 139 patients in the Anakinra group in the study. Development of any thromboembolic event (5% vs 12.3%, p = 0.038; OR 4.3) and PTE (2.9% vs 9.6%, p = 0.023; OR 5.1) were lower in the Anakinra group than SoC. No patient experienced cerebrovascular accident and/or clinically evident deep venous thrombosis both in two arms. After 1:1 PS matching, 88 patients in SoC and 88 patients in the Anakinra group were matched and included in the analysis. In survival analysis, the development of any thromboembolic event, pulmonary thromboembolism, and acute coronary syndrome (ACS) were higher in SoC compared to Anakinra. Survival rate was also lower in patients with SoC arm than Anakinra in patients who had any thromboembolic event as well as ACS. In our study, the development of thrombosis was associated with hyperinflammation in patients with severe and critical COVID-19. Intravenous high-dose anakinra treatment decreases both venous and arterial events in patients with severe and critical COVID-19.
Collapse
Affiliation(s)
- Ramazan Çakmak
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Istinye University, Istanbul, Turkey
| | - Servet Yüce
- Department of Public Health and Biostatistics, Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Mustafa Ay
- Aksaray University, Aksaray Training and Research Hospital, Aksaray, Turkey
| | | | - Muhammed İkbal Kılıç
- Department of Internal Medicine, Aksaray Training and Research Hospital, Aksaray, Turkey
| | - Murat Bektaş
- Division of Rheumatology, Department of Internal Medicine, Aksaray Training and Research Hospital, Yeni Sanayi Street, Merkez, 68200, Aksaray, Turkey.
- Division of Rheumatology, Department of Internal Medicine, Istanbul Aydın University, Istanbul, Turkey.
| |
Collapse
|
65
|
Othman HY, Zaki IAH, Isa MR, Ming LC, Zulkifly HH. A systematic review of thromboembolic complications and outcomes in hospitalised COVID-19 patients. BMC Infect Dis 2024; 24:484. [PMID: 38730292 PMCID: PMC11088167 DOI: 10.1186/s12879-024-09374-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 05/03/2024] [Indexed: 05/12/2024] Open
Abstract
Thromboembolic (TE) complications [myocardial infarction (MI), stroke, deep vein thrombosis (DVT), and pulmonary embolism (PE)] are common causes of mortality in hospitalised COVID-19 patients. Therefore, this review was undertaken to explore the incidence of TE complications and mortality associated with TE complications in hospitalised COVID-19 patients from different studies. A literature search was performed using ScienceDirect and PubMed databases using the MeSH term search strategy of "COVID-19", "thromboembolic complication", "venous thromboembolism", "arterial thromboembolism", "deep vein thrombosis", "pulmonary embolism", "myocardial infarction", "stroke", and "mortality". There were 33 studies included in this review. Studies have revealed that COVID-19 patients tend to develop venous thromboembolism (PE:1.0-40.0% and DVT:0.4-84%) compared to arterial thromboembolism (stroke:0.5-15.2% and MI:0.8-8.7%). Lastly, the all-cause mortality of COVID-19 patients ranged from 4.8 to 63%, whereas the incidence of mortality associated with TE complications was between 5% and 48%. A wide range of incidences of TE complications and mortality associated with TE complications can be seen among hospitalized COVID-19 patients. Therefore, every patient should be assessed for the risk of thromboembolic complications and provided with an appropriate thromboprophylaxis management plan tailored to their individual needs.
Collapse
Affiliation(s)
- Hanies Yuhana Othman
- Department of Clinical Pharmacy, Fakulti Farmasi, Universiti Teknologi MARA Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam, Selangor, Malaysia
| | - Izzati Abdul Halim Zaki
- Department of Clinical Pharmacy, Fakulti Farmasi, Universiti Teknologi MARA Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam, Selangor, Malaysia
- Cardiology Therapeutics Research Group, Universiti Teknologi MARA, Puncak Alam, Selangor, Malaysia
| | - Mohamad Rodi Isa
- Faculty of Medicine, Universiti Teknologi MARA Selangor, Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia
| | - Long Chiau Ming
- School of Medical and Life Sciences, Sunway University, Sunway City, Selangor, Malaysia
| | - Hanis Hanum Zulkifly
- Department of Clinical Pharmacy, Fakulti Farmasi, Universiti Teknologi MARA Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam, Selangor, Malaysia.
- Cardiology Therapeutics Research Group, Universiti Teknologi MARA, Puncak Alam, Selangor, Malaysia.
| |
Collapse
|
66
|
Singh T, Rao Padubidri J, Shetty PH, Antony Manoj M, Mary T, Thejaswi Pallempati B. The top 50 most-cited articles about COVID-19 and the complications of COVID-19: A bibliometric analysis. F1000Res 2024; 13:105. [PMID: 39149509 PMCID: PMC11325134 DOI: 10.12688/f1000research.145713.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 08/17/2024] Open
Abstract
Background This bibliometric analysis examines the top 50 most-cited articles on COVID-19 complications, offering insights into the multifaceted impact of the virus. Since its emergence in Wuhan in December 2019, COVID-19 has evolved into a global health crisis, with over 770 million confirmed cases and 6.9 million deaths as of September 2023. Initially recognized as a respiratory illness causing pneumonia and ARDS, its diverse complications extend to cardiovascular, gastrointestinal, renal, hematological, neurological, endocrinological, ophthalmological, hepatobiliary, and dermatological systems. Methods Identifying the top 50 articles from a pool of 5940 in Scopus, the analysis spans November 2019 to July 2021, employing terms related to COVID-19 and complications. Rigorous review criteria excluded non-relevant studies, basic science research, and animal models. The authors independently reviewed articles, considering factors like title, citations, publication year, journal, impact factor, authors, study details, and patient demographics. Results The focus is primarily on 2020 publications (96%), with all articles being open access. Leading journals include The Lancet, NEJM, and JAMA, with prominent contributions from Internal Medicine (46.9%) and Pulmonary Medicine (14.5%). China played a major role (34.9%), followed by France and Belgium. Clinical features were the primary study topic (68%), often utilizing retrospective designs (24%). Among 22,477 patients analyzed, 54.8% were male, with the most common age group being 26-65 years (63.2%). Complications of COVID-19 affected 13.9% of patients, with a recovery rate of 57.8%. Conclusion Analyzing these top-cited articles offers clinicians and researchers a comprehensive, timely understanding of influential COVID-19 literature. This approach uncovers attributes contributing to high citations and provides authors with valuable insights for crafting impactful research. As a strategic tool, this analysis facilitates staying updated and making meaningful contributions to the dynamic field of COVID-19 research.
Collapse
Affiliation(s)
- Tanya Singh
- Kasturba Medical College Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Jagadish Rao Padubidri
- Department of Forensic Medicine and Toxicology, Kasturba Medical College Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Pavanchand H Shetty
- Department of Forensic Medicine and Toxicology, Kasturba Medical College Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Matthew Antony Manoj
- Kasturba Medical College Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Therese Mary
- Kasturba Medical College Mangalore, Manipal Academy of Higher Education, Manipal, India
| | | |
Collapse
|
67
|
Zhong L, Lin Q, He L, Liu D, Zhu L, Zeng Q, Song J. Time to maximum amplitude of thromboelastography can predict mortality in patients with severe COVID-19: a retrospective observational study. Front Med (Lausanne) 2024; 11:1356283. [PMID: 38756947 PMCID: PMC11097111 DOI: 10.3389/fmed.2024.1356283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
Objective To predict mortality in severe patients with COVID-19 at admission to the intensive care unit (ICU) using thromboelastography (TEG). Methods This retrospective, two-center, observational study involved 87 patients with PCR-and chest CT-confirmed severe COVID-19 who were admitted to at Wuhan Huoshenshan Hospital and the 908th Hospital of Chinese PLA Logistic Support Force between February 2020 and February 2023. Clinic demographics, laboratory results, and outcomes were compared between those who survived and those who died during hospitalization. Results Thromboelastography showed that of the 87 patients, 14 were in a hypercoagulable state, 25 were in a hypocoagulable state, and 48 were normal, based on the time to maximum amplitude (TMA). Patients who died showed significantly lower α angle, but significantly longer R-time, K-time and TMA than patients who survived. Random forest selection showed that K-time, TMA, prothrombin time (PT), international normalized ratio (INR), D-dimer, C-reactive protein (CRP), aspartate aminotransferase (AST), and total bilirubin (Tbil) were significant predictors. Multivariate logistic regression identified that TMA and CRP were independently associated with mortality. TMA had a greater predictive power than CRP levels based on time-dependent AUCs. Patients with TMA ≥ 26.4 min were at significantly higher risk of mortality (hazard ratio 3.99, 95% Confidence Interval, 1.92-8.27, p < 0.01). Conclusion TMA ≥26.4 min at admission to ICU may be an independent predictor of in-hospital mortality for patients with severe COVID-19.
Collapse
Affiliation(s)
- Lincui Zhong
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
| | - Qingwei Lin
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
| | - Longping He
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
| | - Dongmei Liu
- Intensive Care Unit, Huoshenshan Hospital, Wuhan, Hubei, China
- Intensive Care Unit, The 940th Hospital of Chinese PLA Logistic Support Force, Lanzhou, Gansu, China
| | - Lin Zhu
- Intensive Care Unit, Huoshenshan Hospital, Wuhan, Hubei, China
- Department of Critical Care Medicine, The 944th Hospital of Chinese PLA Logistic Support Force, Jiuquan, Gansu, China
| | - Qingbo Zeng
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
| | - Jingchun Song
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
- Intensive Care Unit, Huoshenshan Hospital, Wuhan, Hubei, China
| |
Collapse
|
68
|
Morikawa K, Tabira K, Takemura H, Inaba S, Suzuki Y, Hataji O. Krebs von den Lungen-6 levels at admission predicts exercise-induced hypoxemia before and after discharge in patients with COVID-19. Respir Investig 2024; 62:369-374. [PMID: 38637059 DOI: 10.1016/j.resinv.2024.02.011] [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: 11/19/2023] [Revised: 02/08/2024] [Accepted: 02/18/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND There are no reports of exercise-induced hypoxemia in patients with coronavirus disease 2019 (COVID-19). Additionally, the predictive factors and prevalence of exercise-induced hypoxemia are unknown. This study investigated the incidence and predictive factors of exercise-induced hypoxemia before and after discharge in patients with COVID-19. METHODS We enrolled 77 patients diagnosed with COVID-19 who were hospitalized between November 2020 and October 2021 and who underwent a 6-min walk test before and after discharge. Based on the test results, we classified patients into exercise-induced and non-exercise-induced hypoxemia groups and investigated the predictive factors of exercise-induced hypoxemia using logistic regression analysis. RESULTS The incidences of exercise-induced hypoxemia in patients with COVID-19 were 37.7% and 19.5% before and after discharge, respectively. At admission, the Krebs von den Lungen-6 levels was the associated factor for exercise-induced hypoxemia in patients with COVID-19 before and after discharge, with cut-off values of 314 U/mL and 367 U/mL, respectively. Age and lactate dehydrogenase levels were the associated factors for exercise-induced hypoxemia in patients with COVID-19 before discharge, with cut-off values of 61 years and 492 U/L, respectively. CONCLUSIONS Some patients with COVID-19 may continue to experience exercise-induced hypoxemia after discharge. Age, lactate dehydrogenase, and Krebs von den Lungen-6 levels at admission could serve as predictive markers of exercise-induced hypoxemia before and after discharge in these patients.
Collapse
Affiliation(s)
- Keisuke Morikawa
- Department of Rehabilitation, Matsusaka Municipal Hospital, Tonomachi 1550, Matsusaka, Mie, 515-8544, Japan; Department of Health Science, Kio University Graduate School, Umamichu 4-2-2, Kitakatsuragigun-koryocho, Nara, 635-0832, Japan.
| | - Kazuyuki Tabira
- Department of Health Science, Kio University Graduate School, Umamichu 4-2-2, Kitakatsuragigun-koryocho, Nara, 635-0832, Japan
| | - Hiroyuki Takemura
- Department of Rehabilitation, Matsusaka Municipal Hospital, Tonomachi 1550, Matsusaka, Mie, 515-8544, Japan
| | - Shogo Inaba
- Department of Rehabilitation, Matsusaka Municipal Hospital, Tonomachi 1550, Matsusaka, Mie, 515-8544, Japan
| | - Yuta Suzuki
- Department of Rehabilitation, Matsusaka Municipal Hospital, Tonomachi 1550, Matsusaka, Mie, 515-8544, Japan
| | - Osamu Hataji
- Department of Respiratory Center, Matsusaka Municipal Hospital, Tonomachi 1550, Matsusaka, Mie, 515-8544, Japan
| |
Collapse
|
69
|
Zekić T, Belančić A. Antiphospholipid syndrome, thrombosis, and vaccination in the COVID-19 pandemic. Rheumatol Int 2024; 44:749-755. [PMID: 38393386 DOI: 10.1007/s00296-023-05531-y] [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: 10/03/2023] [Accepted: 12/29/2023] [Indexed: 02/25/2024]
Abstract
Thrombosis is one of the many signs of antiphospholipid syndrome (APS) and COVID-19 infection. Although the mechanisms contributing to thrombosis in APS and COVID-19 are relatively similar, this remains an open subject. Even now (when the COVID-19 pandemic has subsided), there is no conclusive solution to APS and COVID-19 co-occurrence. The presence of newly generated antiphospholipid antibodies (aPLs) in COVID-19 infection may or may not be connected to the diagnosis of APS. The prevalence of aPLs is substantial in severe COVID-19 but not related to thrombosis or a worse outcome. Adequate monitoring of antibody positivity over time is recommended for APL diagnosis. On the other hand, thrombosis and thrombocytopenia can rarely occur with vaccination with mRNA vaccines. Some studies have shown that COVID-19 immunization is well tolerated among APS patients who are triple-positive for aPL, which may comfort patients and referring physicians and lessen hesitation in unvaccinated APS/aPL-positive patients. In this narrative review, we will give an overview of the interaction between aPL-APS-COVID-19-thrombosis and related diagnostic insights learned during the pandemic.
Collapse
Affiliation(s)
- Tatjana Zekić
- Clinical Hospital Center Rijeka, Department of Rheumatology and Clinical Immunology, University of Rijeka, Faculty of Medicine, Rijeka, Croatia.
- University of Rijeka, Faculty of Medicine, 51000, Rijeka, Croatia.
| | - Andrej Belančić
- Department of Clinical Pharmacology, Clinical Hospital Centre Rijeka, Krešimirova 42, 51000, Rijeka, Croatia
- Department of Basic and Clinical Pharmacology with Toxicology, University of Rijeka, Faculty of Medicine, Braće Branchetta 20, 51000, Rijeka, Croatia
| |
Collapse
|
70
|
Appiah L, Jones S, Misra S. Ischemic Colitis in a Middle-Aged Man With COVID-19: Case Report and Review of Literature. HCA HEALTHCARE JOURNAL OF MEDICINE 2024; 5:125-128. [PMID: 38984219 PMCID: PMC11229593 DOI: 10.36518/2689-0216.1362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Introduction Coronavirus disease 2019 (COVID-19) was a pandemic that began in 2019 and continues to have morbid and deadly consequences throughout the world. During the beginning of the pandemic, many considered older adults and immunocompromised younger adults to be the only populations at risk for the severe consequences of COVID-19. Throughout the pandemic, this was proven wrong with several case reports and studies showing that relatively younger adults can also suffer serious consequences from this perplexing virus. Case Presentation We report a rare case of ischemic colitis in a 42-year-old obese man who presented to the emergency department with quintessential COVID-19 symptoms. During his hospital course, he developed not only respiratory failure but also ischemic colitis, although he had no past medical history of any coagulopathy and was never on any pressors. Conclusion As more case reports are being published, it has become evident that COVID-19 has the ability to cause serious extrapulmonary consequences due to an imposed state of hypercoagulability, and younger adults are at risk of facing these consequences, especially if they are obese. Thus, it is imperative that younger adults seek out the COVID-19 vaccine when available to them not only to protect those most vulnerable around them but also to protect themselves from these complications.
Collapse
|
71
|
Wilson R, Ko C, Kashyap R. COVID-19 in the Setting of HIV and Severe Hemophilia A: A Case Report. HCA HEALTHCARE JOURNAL OF MEDICINE 2024; 5:145-150. [PMID: 38984227 PMCID: PMC11229595 DOI: 10.36518/2689-0216.1615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Introduction A hyperactive immune response is the driving force behind severe Coronavirus disease 2019 (COVID-19). Complications of severe COVID-19 include acute respiratory distress syndrome, acute respiratory failure, and increased risk of venous thromboembolism (VTE). The management of patients with COVID-19 includes respiratory support, decreasing immune response to the virus to prevent the progression of disease, and anticoagulation to prevent VTE. Case Presentation We present the case of a patient with a history of human immunodeficiency virus (HIV) and hemophilia A admitted with COVID-19. This case demonstrates the difficulties present when managing COVID-19 in patients with specific comorbidities. Anticoagulation is a recommended component of COVID-19 treatment but is contraindicated in patients with severe hemophilia due to increased risk for bleeding. Research has also shown that dexamethasone decreases mortality in patients with COVID-19, but doctors should use dexamethasone cautiously in patients with HIV since it is an immunosuppressant. Taking certain antiretroviral therapies, such as rilpivirine, also contraindicates the use of dexamethasone. Conclusion In this case, it is important to monitor for the risk and presence of superimposed bacterial or opportunistic infections. Treating a patient with these comorbidities who is infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) highlights the importance of balancing the risks and benefits.
Collapse
Affiliation(s)
- Ryan Wilson
- TriStar Southern Hills Medical Center, Nashville, TN
| | - Chae Ko
- TriStar Southern Hills Medical Center, Nashville, TN
| | - Rahul Kashyap
- TriStar Southern Hills Medical Center, Nashville, TN
| |
Collapse
|
72
|
Ramirez-Cervantes KL, Campillo-Morales S, García-Poza P, Quintana-Díaz M, Huerta-Álvarez C. Antithrombotic Use Patterns in COVID-19 Patients from Spain: A Real-World Data Study. J Clin Med 2024; 13:2403. [PMID: 38673678 PMCID: PMC11051525 DOI: 10.3390/jcm13082403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Antithrombotics have been widely used to treat and prevent COVID-19-related thrombosis; however, studies on their use at population levels are limited. We aimed to describe antithrombotic use patterns during the pandemic in Spanish primary care and hospital-admitted patients with COVID-19. Methods: A real-world data study was performed. Data were obtained from BIFAP's electronic health records. We investigated the antithrombotic prescriptions made within ±14 days after diagnosis between March 2020 and February 2022, divided their use into prior and new/naive groups, and reported their post-discharge use. Results: We included 882,540 individuals (53.4% women), of whom 78,499 were hospitalized. The median age was 44.7 (IQR 39-59). Antithrombotics were prescribed in 37,183 (4.6%) primary care subjects and 42,041 (53.6%) hospital-admitted patients, of whom 7505 (20.2%) and 20,300 (48.3%), respectively, were naive users. Prior users were older and had more comorbidities than new users. Enoxaparin was the most prescribed antithrombotic in hospitals, with higher prescription rates in new than prior users (2348.2, IQR 2390-3123.1 vs. 1378, IQR 1162-1751.6 prescriptions per 10,000 cases, p = 0.002). In primary care, acetylsalicylic acid was the most used antithrombotic, with higher use rates in prior than in naïve users. Post-discharge use occurred in 6686 (15.9%) subjects (median use = 10 days, IQR 9-30). Conclusions: Our study identified a consensus on prescribing antithrombotics in COVID-19 patients, but with low use rates in hospitals.
Collapse
Affiliation(s)
| | - Salvador Campillo-Morales
- Patient Blood Management Research Group, Hospital La Paz Institute for Health Research, 28040 Madrid, Spain;
| | - Patricia García-Poza
- Division of Pharmacoepidemiology and Pharmacovigilance, Spanish Agency for Medicines and Medical Devices (AEMPS), C/Campezo n° 1, Edificio 8, 28022 Madrid, Spain;
| | - Manuel Quintana-Díaz
- Intensive Care Unit, Patient Blood Management Research Group, Research Institute of La Paz University Hospital, La Paz University Hospital, 28040 Madrid, Spain
| | - Consuelo Huerta-Álvarez
- Department of Public Health and Maternity Childcare, Faculty of Medicine, Complutense University of Madrid, Pl. de Ramón y Cajal, s/n, 28040 Madrid, Spain;
| |
Collapse
|
73
|
Kumar H, Gupta R. Neuroinvasion of severe acute respiratory syndrome corona virus-2 (SARS-CoV-2): future risk of epilepsy. Int J Neurosci 2024; 134:323-332. [PMID: 35815479 DOI: 10.1080/00207454.2022.2100784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/24/2022] [Accepted: 06/30/2022] [Indexed: 10/17/2022]
Abstract
Central nervous system (CNS) infection is one of the important risk factors for epilepsy. COVID-19 pandemic, which is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has primarily been considered to involve respiratory system only, but it can also affect the CNS. A wide range of neurological manifestations have been reported in SARS-CoV-2 infected patients including seizures, status epilepticus, stroke, which are considered as important risk factors for the development of epilepsy. In post-mortem, brain tissue samples of COVID-19 patients have shown neuropathological changes and presence of SARS-CoV-2 RNA and viral proteins. In this review, mechanisms of SARS-CoV-2 neuroinvasion like neuronal retrograde trans-synaptic route and vascular route are described along with important neurological manifestations in COVID-19 patients such as seizures and cerebrovascular diseases, which have been found to be associated with the development of epilepsy. Hence, an increased risk of future burden of epilepsy in susceptible COVID-19 survivors has been proposed and preventive measures are suggested. The present review highlights about the possible association between neurological manifestations and future risk of epilepsy in COVID-19 patients.
Collapse
Affiliation(s)
- Hemant Kumar
- Department of Pharmacology, University College of Medical Sciences, University of Delhi, Delhi, India
| | - Rachna Gupta
- Department of Pharmacology, University College of Medical Sciences, University of Delhi, Delhi, India
| |
Collapse
|
74
|
Naidu AS, Wang CK, Rao P, Mancini F, Clemens RA, Wirakartakusumah A, Chiu HF, Yen CH, Porretta S, Mathai I, Naidu SAG. Precision nutrition to reset virus-induced human metabolic reprogramming and dysregulation (HMRD) in long-COVID. NPJ Sci Food 2024; 8:19. [PMID: 38555403 PMCID: PMC10981760 DOI: 10.1038/s41538-024-00261-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/15/2024] [Indexed: 04/02/2024] Open
Abstract
SARS-CoV-2, the etiological agent of COVID-19, is devoid of any metabolic capacity; therefore, it is critical for the viral pathogen to hijack host cellular metabolic machinery for its replication and propagation. This single-stranded RNA virus with a 29.9 kb genome encodes 14 open reading frames (ORFs) and initiates a plethora of virus-host protein-protein interactions in the human body. These extensive viral protein interactions with host-specific cellular targets could trigger severe human metabolic reprogramming/dysregulation (HMRD), a rewiring of sugar-, amino acid-, lipid-, and nucleotide-metabolism(s), as well as altered or impaired bioenergetics, immune dysfunction, and redox imbalance in the body. In the infectious process, the viral pathogen hijacks two major human receptors, angiotensin-converting enzyme (ACE)-2 and/or neuropilin (NRP)-1, for initial adhesion to cell surface; then utilizes two major host proteases, TMPRSS2 and/or furin, to gain cellular entry; and finally employs an endosomal enzyme, cathepsin L (CTSL) for fusogenic release of its viral genome. The virus-induced HMRD results in 5 possible infectious outcomes: asymptomatic, mild, moderate, severe to fatal episodes; while the symptomatic acute COVID-19 condition could manifest into 3 clinical phases: (i) hypoxia and hypoxemia (Warburg effect), (ii) hyperferritinemia ('cytokine storm'), and (iii) thrombocytosis (coagulopathy). The mean incubation period for COVID-19 onset was estimated to be 5.1 days, and most cases develop symptoms after 14 days. The mean viral clearance times were 24, 30, and 39 days for acute, severe, and ICU-admitted COVID-19 patients, respectively. However, about 25-70% of virus-free COVID-19 survivors continue to sustain virus-induced HMRD and exhibit a wide range of symptoms that are persistent, exacerbated, or new 'onset' clinical incidents, collectively termed as post-acute sequelae of COVID-19 (PASC) or long COVID. PASC patients experience several debilitating clinical condition(s) with >200 different and overlapping symptoms that may last for weeks to months. Chronic PASC is a cumulative outcome of at least 10 different HMRD-related pathophysiological mechanisms involving both virus-derived virulence factors and a multitude of innate host responses. Based on HMRD and virus-free clinical impairments of different human organs/systems, PASC patients can be categorized into 4 different clusters or sub-phenotypes: sub-phenotype-1 (33.8%) with cardiac and renal manifestations; sub-phenotype-2 (32.8%) with respiratory, sleep and anxiety disorders; sub-phenotype-3 (23.4%) with skeleto-muscular and nervous disorders; and sub-phenotype-4 (10.1%) with digestive and pulmonary dysfunctions. This narrative review elucidates the effects of viral hijack on host cellular machinery during SARS-CoV-2 infection, ensuing detrimental effect(s) of virus-induced HMRD on human metabolism, consequential symptomatic clinical implications, and damage to multiple organ systems; as well as chronic pathophysiological sequelae in virus-free PASC patients. We have also provided a few evidence-based, human randomized controlled trial (RCT)-tested, precision nutrients to reset HMRD for health recovery of PASC patients.
Collapse
Affiliation(s)
- A Satyanarayan Naidu
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA.
- N-terminus Research Laboratory, 232659 Via del Rio, Yorba Linda, CA, 92887, USA.
| | - Chin-Kun Wang
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- School of Nutrition, Chung Shan Medical University, 110, Section 1, Jianguo North Road, Taichung, 40201, Taiwan
| | - Pingfan Rao
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- College of Food and Bioengineering, Fujian Polytechnic Normal University, No.1, Campus New Village, Longjiang Street, Fuqing City, Fujian, China
| | - Fabrizio Mancini
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- President-Emeritus, Parker University, 2540 Walnut Hill Lane, Dallas, TX, 75229, USA
| | - Roger A Clemens
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- University of Southern California, Alfred E. Mann School of Pharmacy/D. K. Kim International Center for Regulatory & Quality Sciences, 1540 Alcazar St., CHP 140, Los Angeles, CA, 90089, USA
| | - Aman Wirakartakusumah
- International Union of Food Science and Technology (IUFoST), Guelph, ON, Canada
- IPMI International Business School Jakarta; South East Asian Food and Agriculture Science and Technology, IPB University, Bogor, Indonesia
| | - Hui-Fang Chiu
- Department of Chinese Medicine, Taichung Hospital, Ministry of Health & Well-being, Taichung, Taiwan
| | - Chi-Hua Yen
- Department of Family and Community Medicine, Chung Shan Medical University Hospital; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Sebastiano Porretta
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- President, Italian Association of Food Technology (AITA), Milan, Italy
- Experimental Station for the Food Preserving Industry, Department of Consumer Science, Viale Tanara 31/a, I-43121, Parma, Italy
| | - Issac Mathai
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- Soukya International Holistic Health Center, Whitefield, Bengaluru, India
| | - Sreus A G Naidu
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- N-terminus Research Laboratory, 232659 Via del Rio, Yorba Linda, CA, 92887, USA
| |
Collapse
|
75
|
Zhang Q, Pavlinov I, Ye Y, Zheng W. Therapeutic development targeting host heparan sulfate proteoglycan in SARS-CoV-2 infection. Front Med (Lausanne) 2024; 11:1364657. [PMID: 38618194 PMCID: PMC11014733 DOI: 10.3389/fmed.2024.1364657] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/18/2024] [Indexed: 04/16/2024] Open
Abstract
The global pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to an urgent need for effective therapeutic options. SARS-CoV-2 is a novel coronavirus responsible for the COVID-19 pandemic that has resulted in significant morbidity and mortality worldwide. The virus is known to enter host cells by binding to the angiotensin-converting enzyme 2 (ACE2) receptor, and emerging evidence suggests that heparan sulfate proteoglycans (HSPGs) play a crucial role in facilitating this process. HSPGs are abundant cell surface proteoglycan present in many tissues, including the lung, and have been shown to interact directly with the spike protein of SARS-CoV-2. This review aims to summarize the current understanding of the role of HSPGs in SARS-CoV-2 infection and the potential of developing new therapies targeting HSPGs.
Collapse
Affiliation(s)
- Qi Zhang
- Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Ivan Pavlinov
- Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Wei Zheng
- Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
76
|
Jahanshahi F, Jazayeri SB, Eraghi MM, Reis LO, Hamidikia M, Amiri S, Aghamir SMK. A narrative review on adverse drug reactions of COVID-19 treatments on the kidney. Open Med (Wars) 2024; 19:20230867. [PMID: 38584847 PMCID: PMC10996932 DOI: 10.1515/med-2023-0867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 11/01/2022] [Accepted: 11/18/2023] [Indexed: 04/09/2024] Open
Abstract
Studies showed that the respiratory is not the only system affected by coronavirus 2, while cardiovascular, digestive, and nervous systems, as well as essential organs such as the kidneys, can be affected by this virus. In this review, we have studied the epidemiology, clinical, and laboratory findings on COVID-19 infection renal involvement, mortality, physiopathology, remaining renal sequels after recovery, underlying renal disease, and renal injury due to its treatment. Also, protective measures for kidney injury are explained in three levels. Evidence of viral particles and genome in the urine and renal tubular cells and signs of damage such as microangiopathy, hypercoagulopathy, and fibrosis are found in COVID-19 patients. The result of this study showed, in hospitalized COVID-19 patients, that the rate of acute kidney injury (AKI) was up to 46%, with a mortality ranging from 11 to 96%. A considerable proportion of patients with AKI would remain on renal replacement therapy. Proteinuria and hematuria are observed in 87 and 75% patients, and increased Cr and glomerular filtration rate (GFR) <60 ml/min per 1.73 m2 are observed in 29.6 and 35.3% of the patients, respectively. Remedsivir is considered to have adverse effects on GFR. COVID-19 patients need special attention to prevent AKI. Those with underlying chronic kidney disease or AKI need proper and explicit evaluation and treatment to improve their prognosis and decrease mortality, which should not be limited to the hospitalization period.
Collapse
Affiliation(s)
- Fatemeh Jahanshahi
- Research Committee Member, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Urology Research Center, Tehran University of Medical Sciences, Sina Hospital, Tehran, Iran
| | - Seyed Behnam Jazayeri
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mirahmadi Eraghi
- Urology Research Center, Tehran University of Medical Sciences, Sina Hospital, Tehran, Iran
- School of Medicine, Qeshm International Branch, Islamic Azad University, Qeshm, Iran
| | - Leonardo Oliveira Reis
- UroScience and Department of Surgery (Urology), School of Medical Sciences, University of Campinas, Unicamp, and Pontifical Catholic University of Campinas, PUC-Campinas, Campinas, São Paulo, Brazil
| | - Mahtab Hamidikia
- Research Committee Member, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Amiri
- Rasool Akram Medical Complex, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
77
|
Youness M, Mansour S, Sakr F, Olabi S, Atwi S, Martinez IY, El Khatib S, Hallit S, Salameh P, Malaeb D, Hosseini H. Odds and associated factors for thrombosis development among Lebanese COVID-19 patients: a case-control retrospective study. J Pharm Policy Pract 2024; 17:2319743. [PMID: 38505825 PMCID: PMC10950289 DOI: 10.1080/20523211.2024.2319743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024] Open
Abstract
Background Thromboembolism is reported to be up to 27% in COVID-19 patients due to SARS-CoV-2 infection. Dysregulated systemic inflammation and various patient traits play a vital role in thrombosis progression. Purpose To assess odds and associated factors for thrombosis development among Lebanese COVID-19 patients. Methods This was a case-control retrospective study conducted in January-May 2021. Patients infected with COVID-19 and developed thrombosis were classified as cases and patients who were thrombosis-free identified as control. A questionnaire assessed socio-demographics, clinical parameters, and WHO COVID-19 disease severity. Results Among 267 patients, 26 (9.7%) developed thrombosis and the majority of thrombosis 34.6% was myocardial infarction, and the least (3.8%) was for catheter-related thrombosis. Results showed that the risk of thrombosis development is higher in patients with previous thromboembolic event (OR = 9.160) and previous intake of anti-hypertensive medications at home (OR = 3.116). However, females (OR = 0.330; CI: 0.118-0.925), intake of anticoagulants during hospital admission (OR = 0.126; CI: 0.053-0.300) and non-severe COVID-19 were at lower thrombosis risk (OR = 0.273). Patients who developed thromboembolic events had longer hospital stay (OR = 0.077). Conclusion Patients with COVID-19 and thromboembolism were at higher risk of mortality as compared to patients with COVID-19 but without thromboembolism. The use of anticoagulants significantly reduced the risk for thromboembolism.
Collapse
Affiliation(s)
- Mahmoud Youness
- Research Department, Beirut Cardiac Institute, Beirut, Lebanon
| | - Sara Mansour
- School of Pharmacy, Lebanese International University, Beirut, Lebanon
| | - Fouad Sakr
- School of Pharmacy, Lebanese International University, Beirut, Lebanon
| | - Samer Olabi
- Rafic Hariri University Hospital, Beirut, Lebanon
| | - Sarah Atwi
- Rafic Hariri University Hospital, Beirut, Lebanon
| | | | - Sami El Khatib
- Department of Biomedical Sciences, Lebanese International University, Bekaa, Lebanon
- Center for Applied Mathematics and Bioinformatics (CAMB), Gulf University for Science and Technology, Mubarak Al-Abdullah, Kuwait
| | - Souheil Hallit
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik, Jounieh, Lebanon
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
| | - Pascale Salameh
- INSPECT-LB: Institut National de Santé Publique, Epidémiologie Clinique et Toxicologie, Beirut, Lebanon
- Faculty of Pharmacy, Lebanese University, Hadath, Lebanon
- Department of Primary Care and Population Health, University of Nicosia Medical School, Nicosia, Cyprus
- School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Diana Malaeb
- College of Pharmacy, Gulf Medical University, Ajman, United Arab Emirates
| | - Hassan Hosseini
- Neurology Department, Henri Mondor Hospital, AP-HP, Creteil, France
- UPEC-University Paris-Est, Creteil, France
- RAMSAY SANTÉ, HPPE, Champigny sur Marne, France
| |
Collapse
|
78
|
Alsayed AR, Ahmed SI, AL Shweiki AO, Al-Shajlawi M, Hakooz N. The laboratory parameters in predicting the severity and death of COVID-19 patients: Future pandemic readiness strategies. BIOMOLECULES & BIOMEDICINE 2024; 24:238-255. [PMID: 37712883 PMCID: PMC10950347 DOI: 10.17305/bb.2023.9540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/30/2023] [Accepted: 09/14/2023] [Indexed: 09/16/2023]
Abstract
The range of clinical manifestations associated with the infection by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) encompasses a broad spectrum, ranging from flu-like symptoms to the occurrence of multiple organ failure and death. The severity of the coronavirus disease 2019 (COVID-19) is categorized based on clinical presentation and is divided into three distinct levels of severity identified as non-severe, severe, and critical. Although individuals of all age groups are susceptible to SARS-CoV-2 infection, middle-aged and older adults are more frequently impacted, with the latter being more likely to develop severe illness. Various laboratory characteristics observed in hospitalized COVID-19 patients have been correlated with adverse outcomes. These include elevated levels of D-dimer, liver enzymes, lactate dehydrogenase, C-reactive protein, ferritin, prothrombin time, and troponin, as well as decreased lymphocyte and platelets counts. This review investigated the relationship between baseline clinical characteristics, initial laboratory parameters upon hospital admission, and the severity of illness and mortality rates among COVID-19 patients. Although the COVID-19 pandemic has concluded, understanding the laboratory predictors of virus severity and mortality remains crucial, and examining these predictors can have long-term effects. Such insights can help healthcare systems manage resources more effectively and deliver timely and appropriate care by identifying and targeting high-risk individuals. This knowledge can also help us better prepare for future pandemics. By examining these predictors, we can take steps to protect public health and mitigate the impact of future pandemics.
Collapse
Affiliation(s)
- Ahmad R Alsayed
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Syed Imran Ahmed
- College of Health and Science, School of Pharmacy, University of Lincoln, Lincoln, United Kingdom
| | - Anas Osama AL Shweiki
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Mustafa Al-Shajlawi
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Nancy Hakooz
- School of Pharmacy, The University of Jordan, Amman, Jordan
| |
Collapse
|
79
|
Saito Z, Uchiyama S, Nishioka S, Tamura K, Tamura N, Kuwano K. Predictors of in-hospital mortality in elderly unvaccinated patients during SARS-CoV-2 Alpha variants epidemic. Infect Prev Pract 2024; 6:100341. [PMID: 38357519 PMCID: PMC10864849 DOI: 10.1016/j.infpip.2024.100341] [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: 07/13/2023] [Accepted: 11/21/2023] [Indexed: 02/16/2024] Open
Abstract
Background COVID-19, caused by SARS-CoV-2, has caused a global pandemic. This study aimed to identify predictors of in-hospital mortality in unvaccinated elderly patients with COVID-19 by comparing various predictive factors between the survivors and non-survivors. Methods We retrospectively selected 132 unvaccinated patients aged over 65 years with COVID-19 at a hospital in Kanagawa, Japan, during SARS-CoV-2 Alpha variants epidemic. We compared the clinical characteristics, laboratory and radiological findings, treatment, and complications of the survivors and non-survivors. In logistic regression analysis, variables that were significant in the univariate analysis were subjected to multivariate analysis using the variable increase method. Results There were 119 and 13 patients in the survivor and non-survivor groups, respectively. Multivariate regression revealed increasing odds with the presence of ARDS and DIC (odd ratio (OR) = 16.35, 34.36; P=0.002, 0.001, respectively) and prolonged hospital stay (OR = 1.17; P=0.004). Conclusions We found the complications of ARDS and DIC and hospital length of stay to be independent predictors of in-hospital mortality in elderly unvaccinated patients with COVID-19. Establishing treatments and prevention methods for ARDS and DIC could result in lower mortality rates.
Collapse
Affiliation(s)
- Zenya Saito
- Division of Respiratory Diseases, Department of Internal Medicine, Atsugi City Hospital, Kanagawa, Japan
| | - Shota Uchiyama
- Division of Respiratory Diseases, Department of Internal Medicine, Atsugi City Hospital, Kanagawa, Japan
| | - Saiko Nishioka
- Division of Respiratory Diseases, Department of Internal Medicine, Atsugi City Hospital, Kanagawa, Japan
| | - Kentaro Tamura
- Division of Respiratory Diseases, Department of Internal Medicine, Atsugi City Hospital, Kanagawa, Japan
| | - Nobumasa Tamura
- Division of Respiratory Diseases, Department of Internal Medicine, Atsugi City Hospital, Kanagawa, Japan
| | - Kazuyoshi Kuwano
- Division of Respiratory Diseases, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
80
|
Zou Y, Qin C, Yang Q, Lang Y, Liu K, Yang F, Li X, Zhao Y, Zheng T, Wang M, Shi R, Yang W, Zhou Y, Chen L, Liu F. Clinical characteristics, outcomes and risk factors for mortality in hospitalized diabetes and chronic kidney disease patients after COVID-19 infection following widespread vaccination. J Endocrinol Invest 2024; 47:619-631. [PMID: 37725309 DOI: 10.1007/s40618-023-02180-7] [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: 03/22/2023] [Accepted: 08/17/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND COVID-19 poses a significant threat to patients with comorbidities, such as diabetes and chronic kidney disease (CKD). China experienced a nationwide COVID-19 endemic from December 2022 to January 2023, which is the first occurrence of such an outbreak following China's widespread administration of COVID-19 vaccinations. METHODS A total of 338 patients with diabetes and CKD combined with COVID-19 infection between December 7, 2022 and January 31, 2023 were included in this study. The end follow-up date was February 10, 2023. Univariate analysis and multivariate Cox analysis were used to analyze risk factors for death. RESULTS During the 50-day median follow-up period, 90 patients in the study cohort died, for a mortality rate of 26.63%. The median age of the study cohort was 74 years, with a male predominance of 74%. During hospitalization, 21% of patients had incident AKI, 17% of patients experienced stroke, and 40% of patients experienced respiratory failure. Cox proportional hazard regression showed that older age, a diagnosis of severe or critically severe COVID-19 infection, incident AKI and respiratory failure, higher level of average values of fasting glucose during hospitalization, UA, and total bilirubin were independent risk factors for death in our multivariate model. CONCLUSIONS These findings highlight the critical importance of identifying and managing comorbid risk factors for COVID-19, especially among the elderly, in order to optimize clinical outcomes, even after COVID-19 vaccination.
Collapse
Affiliation(s)
- Y Zou
- Division of Nephrology, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan Province, China
| | - C Qin
- Division of Nephrology, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan Province, China
| | - Q Yang
- Division of Nephrology, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan Province, China
| | - Y Lang
- Division of Nephrology, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan Province, China
| | - K Liu
- Division of Nephrology, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan Province, China
| | - F Yang
- Department of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - X Li
- Department of Clinical Research Management, West China Hospital of Sichuan University, Chengdu, China
| | - Y Zhao
- Department of Clinical Research Management, West China Hospital of Sichuan University, Chengdu, China
| | - T Zheng
- Information Center, West China Hospital of Sichuan University, Chengdu, China
- Engineering Research Center of Medical Information Technology, Ministry of Education, Chengdu, China
| | - M Wang
- Information Center, West China Hospital of Sichuan University, Chengdu, China
- Engineering Research Center of Medical Information Technology, Ministry of Education, Chengdu, China
| | - R Shi
- Information Center, West China Hospital of Sichuan University, Chengdu, China
- Engineering Research Center of Medical Information Technology, Ministry of Education, Chengdu, China
| | - W Yang
- Division of Project Design and Statistics, West China Hospital of Sichuan University, Chengdu, China
| | - Y Zhou
- Integrated Care Management Center, West China Hospital of Sichuan University, Chengdu, China
| | - L Chen
- Department of Clinical Research Management, West China Hospital of Sichuan University, Chengdu, China
- Division of Neurology, West China Hospital of Sichuan University, Chengdu, China
| | - Fang Liu
- Division of Nephrology, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan Province, China.
- Department of Clinical Research Management, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
81
|
Waleed MS, Dhulipalla L, Niazi M, Terjanian T, Dhar M. COVID-Induced Thrombotic Thrombocytopenic Purpura: A Case Report and Treatment-Focused Review. Cureus 2024; 16:e57252. [PMID: 38686279 PMCID: PMC11057214 DOI: 10.7759/cureus.57252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2024] [Indexed: 05/02/2024] Open
Abstract
Thrombotic thrombocytopenic purpura (TTP) is a rare disease that is part of a vast spectrum of thrombotic microangiopathies (TMAs). Despite the rarity of TTP, clinicians must maintain a high suspicion of this disease. The condition is characterized by fever, low platelets, hemolytic anemia, renal abnormalities, and neurological dysfunction. However, all these symptoms are not necessarily present in all the patients. In this review, we describe a case of a 51-year-old female who presented to the emergency department (ED) with chief complaints of dizziness and lightheadedness, subsequently leading to a diagnosis of TTP, caused as a result of COVID-19. This review raises awareness so that there is early recognition of any hematological manifestations associated with COVID-19, reducing the morbidity and mortality associated with the disease. Due to the unpredictability of COVID-19 and its complications, robust research is needed to understand the mechanism and determine which patients are more at risk for adverse outcomes.
Collapse
Affiliation(s)
| | | | - Muhammad Niazi
- Internal Medicine, Northwell Health/Staten Island University Hospital, Staten Island, USA
| | - Terenig Terjanian
- Hematology and Oncology, Staten Island University Hospital, Staten Island, USA
| | - Meekoo Dhar
- Hematology and Oncology, Staten Island University Hospital, Staten Island, USA
| |
Collapse
|
82
|
Kalinin RE, Suchkov IA, Agapov AB, Povarov VO, Mzhavanadze ND. Features of various options for anticoagulant therapy for a new coronavirus infection against the background of obesity. KAZAN MEDICAL JOURNAL 2024; 105:14-25. [DOI: 10.17816/kmj159398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
BACKGROUND: When treating a new coronavirus infection, the prevention of venous thromboembolic complications in obese patients is of particular importance.
AIM: To analyze various options for anticoagulant therapy in obese and non-obese patients in the treatment of a new coronavirus infection.
MATERIAL AND METHODS: The patients were divided into two groups: the first group — obese patients — 151 (40.8%) people [average age 63 (56–69) years], the second group without obesity — 219 (59.2%) people [average age 63 (51.5–71.0) years], p=0.998. According to the prescribed anticoagulant and the presence of obesity, patients were divided into subgroups: subgroup 1 — non-obese + low-molecular-weight heparin (n=114), subgroup 2 — non-obese + unfractionated heparin (n=58), subgroup 3 — obesity + low-molecular-weight heparin (n=76), subgroup 4 — obesity + unfractionated heparin (n=66). Venous thrombotic complications and bleeding rates were assessed. Analysis of qualitative indicators was performed using Pearson χ2 and Fisher tests. The distribution of quantitative indicators was assessed using the Kolmogorov–Smirnov and Shapiro–Wilk tests (p 0.05). Due to the non-normal distribution of indicators, mean values were presented as median (Me) and interquartile range (Q1–Q3), and analysis was carried out using Wilcoxon, Mann–Whitney and Kruskal–Wallis tests.
RESULTS: In nonobese patients, a comparable incidence of deep vein thrombosis was noted on low molecular weight heparin and unfractionated heparin — 1 (0.9%) case versus 3 (5.2%) cases (p=0.102). At the same time, there was a statistically significant difference in the frequency of pulmonary embolism without a source according to autopsy data — 2 (1.8%) versus 4 (6.9%) cases (p=0.004). In obese patients receiving unfractionated heparin, the incidence of this complication was higher and amounted to 9 (13.6%), and in patients receiving low molecular weight heparin — 2 (2.6%) cases (p=0.004). A higher incidence of bleeding was observed in patients with obesity compared to patients without it (10.6 vs 4.7% of cases, p=0.045). When analyzing hemorrhagic complications, it was found that the incidence of major and significant bleeding was higher in patients receiving unfractionated heparin rather than low molecular weight heparin (16.7 vs 5.3% of cases, p 0.001).
CONCLUSION: The use of low molecular weight heparin in obese patients with new coronavirus infection was associated with a low incidence of pulmonary embolism and bleeding compared with patients receiving unfractionated heparin.
Collapse
|
83
|
Gu J, Wang Y, Zhang JF, Wang CQ. The impacts of prophylactic anticoagulation therapy during hospitalization on long-term cardiovascular outcomes in high-risk COVID-19 patients amid the omicron wave of the pandemic. IJC HEART & VASCULATURE 2024; 50:101353. [PMID: 38347941 PMCID: PMC10859301 DOI: 10.1016/j.ijcha.2024.101353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 01/30/2024] [Indexed: 02/15/2024]
Abstract
Background Although prophylactic anticoagulation therapy is suggested to be adopted in severe COVID-19 patients, its effects on the long-term cardiovascular (CV) outcomes, namely the risk of major adverse CV events(MACEs) in high-risk CV patients amid the omicron wave of the pandemic, remain unknown. Methods We conducted this prospective cohort study of consecutive adults hospitalized COVID-19 between 19 April and 12 June 2022, COVID-19 patients with at least two CV risk factors or pre-existing CV diseases were enrolled. A propensity score matching(PSM) method was used to evaluated the effects of prophylactic anticoagulation therapy in hospital on long-term MACEs, including CV death, non-fatal myocardial infarction, non-fatal stroke, hospitalization due to unstable angina pectoris, coronary revascularization and arterial or venous thrombosis. Results Two cohorts (with or without anticoagulants during hospitalization) of each 230 patients with balanced baseline characteristics were formed using PSM. During the 15-month follow-up period, 13 patients with anticoagulants and 29 patients without anticoagulants developed MACEs. Overall, the anticoagulation group had a significantly lower risk of MACEs than the control group (hazard ratio [HR] 0.431; 95 % confidence interval [CI]: 0.224-0.830, P = 0.010). Regarding specific constituents of MACEs, the differences were mainly reflected in arterial or venous thrombosis. The significantly lower HRs of overall MACEs were significantly observed in subgroup of age > 75 years, women, higher D dimer level, unvaccinated and non-nirmatrelvir-ritonavir prescribed patients. Conclusions Prophylactic anticoagulation therapy during hospitalization was effective in reducing long-term MACEs among COVID-19 patients with CV risk factors or pre-existing CV diseases amid the omicron wave of the pandemic.
Collapse
Affiliation(s)
- Jun Gu
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Yue Wang
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Jun-feng Zhang
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Chang-qian Wang
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
84
|
Alana NB, Ciurylo WA, Hurlock N. HMG-CoA reductase inhibitors and the attenuation of risk for disseminated intravascular coagulation in patients with sepsis. J Thromb Thrombolysis 2024; 57:260-268. [PMID: 37945940 DOI: 10.1007/s11239-023-02910-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/22/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Disseminated Intravascular Coagulation (DIC) is a syndrome of dysregulated coagulation. Patients with sepsis are at increased risk for DIC. HMG-CoA Reductase Inhibitors (Statins) are primarily used as lipid-lowering agents; however, studies have suggested statins may possess anti-inflammatory, antithrombotic, anticoagulant, and endothelial stabilizing properties. These mechanisms may oppose those that underlie the pathogenesis of septic DIC. METHODS To evaluate whether statins may be protective against the development of DIC, we conducted a multi-center, retrospective case-control study where 86,638 critically ill patients admitted to the ICU with sepsis, severe sepsis or septic shock were identified during a 3-year period. Patients who developed DIC during their hospitalization were identified and stratified by whether they received a statin or not during their hospitalization. Odds ratios for development of DIC was calculated by composite of any statin, as well as low, moderate, and high intensity statins. RESULTS 2236 patients would develop DIC compared to 84,402 who did not. The use of any statin was associated with a reduced likelihood for developing DIC (odds ratio [OR], 0.69; 95% CI, 0.61-0.78). This was observed with use of both moderate (OR, 0.64; 95% CI, 0.53-0.77) and high (OR, 0.72; 95% CI, 0.61-0.84) but not low intensity statins (OR, 0.84; 95% CI, 0.53-1.32). CONCLUSIONS The use of moderate and high intensity statins was associated with a significantly reduced odds of developing DIC in critically ill patients with sepsis. This present study may be the first to suggest that statin medications may independently reduce the frequency of DIC in critically ill patients with severe sepsis or septic shock. More research is needed to investigate the potential for this class of medication to be protective against DIC.
Collapse
Affiliation(s)
- Nicholas B Alana
- Portsmouth Regional Hospital Internal Medicine Residency Program, HCA Healthcare/Tufts University School of Medicine, 333 Borthwick Ave. Portsmouth, New Hampshire, 03801, USA.
| | - William A Ciurylo
- Portsmouth Regional Hospital Internal Medicine Residency Program, HCA Healthcare/Tufts University School of Medicine, 333 Borthwick Ave. Portsmouth, New Hampshire, 03801, USA
| | - Natalie Hurlock
- HCA Healthcare, 2000 Health Park Drive, Brentwood, TN, 37027, UK
| |
Collapse
|
85
|
Mansour HM. The interference between SARS-COV-2 and Alzheimer's disease: Potential immunological and neurobiological crosstalk from a kinase perspective reveals a delayed pandemic. Ageing Res Rev 2024; 94:102195. [PMID: 38244862 DOI: 10.1016/j.arr.2024.102195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/22/2024]
Abstract
Coronavirus disease 2019 (COVID-19) has infected over 700 million people, with up to 30% developing neurological manifestations, including dementias. However, there is a lack of understanding of common molecular brain markers causing Alzheimer's disease (AD). COVID-19 has etiological cofactors with AD, making patients with AD a vulnerable population at high risk of experiencing more severe symptoms and worse consequences. Both AD and COVID-19 have upregulated several shared kinases, leading to the repositioning of kinase inhibitors (KIs) for the treatment of both diseases. This review provides an overview of the interactions between the immune system and the nervous system in relation to receptor tyrosine kinases, including epidermal growth factor receptors, vascular growth factor receptors, and non-receptor tyrosine kinases such as Bruton tyrosine kinase, spleen tyrosine kinase, c-ABL, and JAK/STAT. We will discuss the promising results of kinase inhibitors in pre-clinical and clinical studies for both COVID-19 and Alzheimer's disease (AD), as well as the challenges in repositioning KIs for these diseases. Understanding the shared kinases between AD and COVID-19 could help in developing therapeutic approaches for both.
Collapse
Affiliation(s)
- Heba M Mansour
- General Administration of Innovative Products, Central Administration of Biological, Innovative Products, and Clinical Studies (Bio-INN), Egyptian Drug Authority (EDA), Giza, Egypt.
| |
Collapse
|
86
|
Connors JM, Ariëns RAS. Uncertainties about the roles of anticoagulation and microclots in post-acute sequelae of SARS-CoV-2 infection-response to the letter by Kell et al. J Thromb Haemost 2024; 22:569-571. [PMID: 38309814 DOI: 10.1016/j.jtha.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 11/11/2023] [Indexed: 02/05/2024]
Affiliation(s)
- Jean M Connors
- Hematology Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK. https://twitter.com/RobertAriens
| |
Collapse
|
87
|
Kell DB, Khan MA, Laubscher GJ, Pretorius E. Uncertainties about the roles of anticoagulation and microclots in postacute sequelae of SARS-CoV-2 infection: comment from Kell et al. J Thromb Haemost 2024; 22:565-568. [PMID: 38309813 DOI: 10.1016/j.jtha.2023.09.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/14/2023] [Accepted: 09/29/2023] [Indexed: 02/05/2024]
Affiliation(s)
- Douglas B Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom; The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark; Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.
| | - M Asad Khan
- North West Lung Centre, Manchester University Hospitals, Manchester, United Kingdom
| | | | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom; Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
88
|
Haile AT, Haile RG, Gebrehiwot EH, Abeje EW. Venous Thromboembolism and Associated Factors in Hospitalized Patients with COVID-19 at Addis Ababa COVID-19 Field Hospital, Ethiopia. Infect Drug Resist 2024; 17:305-317. [PMID: 38293313 PMCID: PMC10826547 DOI: 10.2147/idr.s449401] [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: 12/11/2023] [Accepted: 01/19/2024] [Indexed: 02/01/2024] Open
Abstract
Background The association of COVID-19 with venous thromboembolism is of particular interest as there are reports that have associated thrombotic events with the pandemic. COVID-19 may predispose to venous thromboembolism. There is initial evidence suggesting that individuals with COVID-19 may be more prone to developing venous thromboembolism (VTE). This study aims to assess venous thromboembolism and associated factors in 19 patients admitted to Addis Ababa's field hospital for COVID-19. Objective To identify associated factors that affect the development of thromboembolism in patients admitted with the diagnosis of COVID-19. Methods To determine the risks of VTE patients, an institution-based case control research was conducted using SPSS version 26, multivariable binary logistic regression analysis was used. Results Significant factors associated with the development of VTE in COVID-19 patients included having a severe case (AOR = 0.38, 95% CI = 0.149-0.961), risk factors for VTE (AOR = 2.57, 95% CI = 1.18-5.33), diabetes (AOR = 3.745, 95% CI = 1.715-8.176), chest pain (AOR = 4.13, 95% CI = 1.89-9), stage 1 hypertension levels (AOR = 3.67, 95% CI = 1.37-9.836), and hospital anticoagulation (AOR = 11.78, 95% CI = 5.25-26.4). Conclusion The results of VTE in individuals with COVID-19 imply a direct association between severe COVID-19 and diabetes, having risk factors, hypertension, and hospital anticoagulation.
Collapse
Affiliation(s)
- Abel Teklit Haile
- Research Department, Saint Peter Specialized Hospital, Addis Ababa, Ethiopia
- Department of Internal Medicine, Wollo University, Dessie, Ethiopia
| | - Robel Gemechu Haile
- Research Department, Saint Peter Specialized Hospital, Addis Ababa, Ethiopia
- Department of Internal Medicine, Wollo University, Dessie, Ethiopia
- School of Public Health, Gamby Medical and Business College, Addis Ababa, Ethiopia
| | - Esrom Hagos Gebrehiwot
- Research Department, Saint Peter Specialized Hospital, Addis Ababa, Ethiopia
- Department of Internal Medicine, Wollo University, Dessie, Ethiopia
- School of Public Health, Gamby Medical and Business College, Addis Ababa, Ethiopia
| | - Eden Workalemahu Abeje
- Research Department, Saint Peter Specialized Hospital, Addis Ababa, Ethiopia
- Department of Internal Medicine, Wollo University, Dessie, Ethiopia
- School of Public Health, Gamby Medical and Business College, Addis Ababa, Ethiopia
| |
Collapse
|
89
|
Mackiewicz-Milewska M, Cisowska-Adamiak M, Pyskir J, Świątkiewicz I. Venous Thromboembolism in Patients Hospitalized for COVID-19 in a Non-Intensive Care Unit. J Clin Med 2024; 13:528. [PMID: 38256663 PMCID: PMC10816041 DOI: 10.3390/jcm13020528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
Coronavirus Disease 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) may contribute to venous thromboembolism (VTE) with adverse effects on the course of COVID-19. The purpose of this study was to investigate an incidence and risk factors for VTE in patients hospitalized for COVID-19 in a non-intensive care unit (non-ICU). Consecutive adult patients with COVID-19 hospitalized from November 2021 to March 2022 in the isolation non-ICU at our center were included in the study. Incidence of VTE including pulmonary embolism (PE) and deep vein thrombosis (DVT), clinical characteristics, and D-dimer plasma levels during the hospitalization were retrospectively evaluated. Among the 181 patients (aged 68.8 ± 16.2 years, 44% females, 39% Delta SARS-CoV-2 variant, 61% Omicron SARS-CoV-2 variant), VTE occurred in 29 patients (VTE group, 16% of the entire cohort). Of them, PE and DVT were diagnosed in 15 (8.3% of the entire cohort) and 14 (7.7%) patients, respectively. No significant differences in clinical characteristics were observed between the VTE and non-VTE groups. On admission, median D-dimer was elevated in both groups, more for VTE group (1549 ng/mL in VTE vs. 1111 ng/mL in non-VTE, p = 0.09). Median maximum D-dimer was higher in the VTE than in the non-VTE group (5724 ng/mL vs. 2200 ng/mL, p < 0.005). In the univariate analysis, systemic arterial hypertension and the need for oxygen therapy were predictors of VTE during hospitalization for COVID-19 (odds ratio 2.59 and 2.43, respectively, p < 0.05). No significant associations were found between VTE risk and other analyzed factors; however, VTE was more likely to occur in patients with a history of VTE, neurological disorders, chronic pulmonary or kidney disease, atrial fibrillation, obesity, and Delta variant infection. Thromboprophylaxis (83.4% of the entire cohort) and anticoagulant treatment (16.6%) were not associated with a decreased VTE risk. The incidence of VTE in patients hospitalized in non-ICU for COVID-19 was high despite the common use of thromboprophylaxis or anticoagulant treatment. A diagnosis of arterial hypertension and the need for oxygen therapy were associated with an increased VTE risk. Continuous D-dimer monitoring is required for the early detection of VTE.
Collapse
Affiliation(s)
- Magdalena Mackiewicz-Milewska
- Department of Rehabilitation, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland;
| | - Małgorzata Cisowska-Adamiak
- Department of Rehabilitation, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland;
| | - Jerzy Pyskir
- Department of Biophysics, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland;
| | - Iwona Świątkiewicz
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA 92037, USA;
| |
Collapse
|
90
|
Velásquez PA, Hernandez JC, Galeano E, Hincapié-García J, Rugeles MT, Zapata-Builes W. Effectiveness of Drug Repurposing and Natural Products Against SARS-CoV-2: A Comprehensive Review. Clin Pharmacol 2024; 16:1-25. [PMID: 38197085 PMCID: PMC10773251 DOI: 10.2147/cpaa.s429064] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 11/14/2023] [Indexed: 01/11/2024] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a betacoronavirus responsible for the COVID-19 pandemic, causing respiratory disorders, and even death in some individuals, if not appropriately treated in time. To face the pandemic, preventive measures have been taken against contagions and the application of vaccines to prevent severe disease and death cases. For the COVID-19 treatment, antiviral, antiparasitic, anticoagulant and other drugs have been reused due to limited specific medicaments for the disease. Drug repurposing is an emerging strategy with therapies that have already tested safe in humans. One promising alternative for systematic experimental screening of a vast pool of compounds is computational drug repurposing (in silico assay). Using these tools, new uses for approved drugs such as chloroquine, hydroxychloroquine, ivermectin, zidovudine, ribavirin, lamivudine, remdesivir, lopinavir and tenofovir/emtricitabine have been conducted, showing effectiveness in vitro and in silico against SARS-CoV-2 and some of these, also in clinical trials. Additionally, therapeutic options have been sought in natural products (terpenoids, alkaloids, saponins and phenolics) with promising in vitro and in silico results for use in COVID-19 disease. Among these, the most studied are resveratrol, quercetin, hesperidin, curcumin, myricetin and betulinic acid, which were proposed as SARS-CoV-2 inhibitors. Among the drugs reused to control the SARS-CoV2, better results have been observed for remdesivir in hospitalized patients and outpatients. Regarding natural products, resveratrol, curcumin, and quercetin have demonstrated in vitro antiviral activity against SARS-CoV-2 and in vivo, a nebulized formulation has demonstrated to alleviate the respiratory symptoms of COVID-19. This review shows the evidence of drug repurposing efficacy and the potential use of natural products as a treatment for COVID-19. For this, a search was carried out in PubMed, SciELO and ScienceDirect databases for articles about drugs approved or under study and natural compounds recognized for their antiviral activity against SARS-CoV-2.
Collapse
Affiliation(s)
- Paula Andrea Velásquez
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Juan C Hernandez
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Elkin Galeano
- Grupo Productos Naturales Marinos, Departamento de Farmacia, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Jaime Hincapié-García
- Grupo de investigación, Promoción y prevención farmacéutica, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia UdeA, Medellín, Colombia
| | - María Teresa Rugeles
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Wildeman Zapata-Builes
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| |
Collapse
|
91
|
Sharathkumar A, Wendt L, Ortman C, Srinivasan R, Chute CG, Chrischilles E, Takemoto CM. COVID-19 outcomes in persons with hemophilia: results from a US-based national COVID-19 surveillance registry. J Thromb Haemost 2024; 22:61-75. [PMID: 37182697 PMCID: PMC10181864 DOI: 10.1016/j.jtha.2023.04.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/29/2023] [Accepted: 04/25/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND Hypercoagulable state contributing to thrombotic complications worsens COVID-19 severity and outcomes, whereas anticoagulation improves outcomes by alleviating hypercoagulability. OBJECTIVES To examine whether hemophilia, an inherent hypocoagulable condition, offers protection against COVID-19 severity and reduces venous thromboembolism (VTE) risk in persons with hemophilia (PwH). METHODS A 1:3 propensity score-matched retrospective cohort study used national COVID-19 registry data (January 2020 through January 2022) to compare outcomes between 300 male PwH and 900 matched controls without hemophilia. RESULTS Analyses of PwH demonstrated that known risk factors (older age, heart failure, hypertension, cancer/malignancy, dementia, and renal and liver disease) contributed to severe COVID-19 and/or 30-day all-cause mortality. Non-central nervous system bleeding was an additional risk factor for poor outcomes in PwH. Odds of developing VTE with COVID-19 in PwH were associated with pre-COVID VTE diagnosis (odds ratio [OR], 51.9; 95% CI, 12.8-266; p < .001), anticoagulation therapy (OR, 12.7; 95% CI, 3.01-48.6; p < .001), and pulmonary disease (OR, 16.1; 95% CI, 10.4-25.4; p < .001). Thirty-day all-cause mortality (OR, 1.27; 95% CI, 0.75-2.11; p = .3) and VTE events (OR, 1.32; 95% CI, 0.64-2.73; p = .4) were not significantly different between the matched cohorts; however, hospitalizations (OR, 1.58; 95% CI, 1.20-2.10; p = .001) and non-central nervous system bleeding events (OR, 4.78; 95% CI, 2.98-7.48; p < .001) were increased in PwH. In multivariate analyses, hemophilia did not reduce adverse outcomes (OR, 1.32; 95% CI, 0.74-2.31; p = .2) or VTE (OR, 1.14; 95% CI, 0.44-2.67; p = .8) but increased bleeding risk (OR, 4.70; 95% CI, 2.98-7.48; p < .001). CONCLUSION After adjusting for patient characteristics/comorbidities, hemophilia increased bleeding risk with COVID-19 but did not protect against severe disease and VTE.
Collapse
Affiliation(s)
- Anjali Sharathkumar
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.
| | - Linder Wendt
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, USA
| | - Chris Ortman
- Department of Bioinformatics, University of Iowa, Iowa City, Iowa, USA; Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Ragha Srinivasan
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Elizabeth Chrischilles
- Department of Bioinformatics, University of Iowa, Iowa City, Iowa, USA; Department of Epidemiology, School of Public Health, University of Iowa, Iowa, USA
| | - Clifford M Takemoto
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
92
|
Zhang Y, Man J, Wang J, Liu J, Song X, Yu X, Li J, Li R, Qiu Y, Li J, Chen Y. Surface modification of polyvinyl chloride with sodium alginate/carboxymethyl chitosan and heparin for realizing the anticoagulation. Int J Biol Macromol 2024; 254:127653. [PMID: 37918597 DOI: 10.1016/j.ijbiomac.2023.127653] [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: 07/02/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
Thrombosis of extracorporeal circuits causes significant morbidity and mortality worldwide. In this study, plasma treatment technology and chemical grafting method were used to construct heparinized surfaces on the PVC substrate, which could not only reduce thrombosis but also decrease the side effects of the direct injection of anticoagulants. The PVC substrate was modified by plasma treatment technology firstly to obtain the active surface with the hydroxyl groups used for grafting. Then, heparin was grafted onto the modified PVC surface using different grafting strategies to prepare different heparinized surfaces. The experimental results indicated that the sodium alginate (SA) and carboxymethyl chitosan (CCS) used as interlayers could significantly increase the graft density of heparin to improve the anticoagulant effects and hemocompatibility of heparinized surfaces. In addition, the modification of heparin can further improve the anticoagulant effects. The CCS/low-molecular-weight heparin (LWMH) surface has the best anticoagulant properties, which can prolong the activated partial thromboplastin time (APTT) values of human plasma for about 35 s, reduce the hemolysis rates to <0.3 %, and perform well in the in-vitro blood circulation test. The heparinized surfaces prepared in this work have great application potential in anticoagulant treatment for medical devices.
Collapse
Affiliation(s)
- Yongqi Zhang
- Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan 250061, PR China; Key National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan 250061, PR China
| | - Jia Man
- Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan 250061, PR China; Key National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan 250061, PR China.
| | - Jiali Wang
- Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Jianing Liu
- Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Xinzhong Song
- Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan 250061, PR China; Key National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan 250061, PR China
| | - Xiaohan Yu
- School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Jianyong Li
- Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan 250061, PR China; Key National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan 250061, PR China
| | - Ruijian Li
- Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Yinghua Qiu
- Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan 250061, PR China; Key National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan 250061, PR China
| | - Jianfeng Li
- Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan 250061, PR China; Key National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan 250061, PR China
| | - Yuguo Chen
- Qilu Hospital of Shandong University, Jinan 250012, PR China
| |
Collapse
|
93
|
Ghavami G, Sardari S. Two Birds with One Stone: Drug Regime Targets Viral Pathogenesis Phases and COVID-19 ARDS at the Same Time. Infect Disord Drug Targets 2024; 24:e290124226467. [PMID: 38288808 DOI: 10.2174/0118715265270637240107153121] [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: 07/23/2023] [Revised: 10/09/2023] [Accepted: 11/20/2023] [Indexed: 09/04/2024]
Abstract
BACKGROUND Severe COVID-19 or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a kind of viral pneumonia induced by infection with the coronavirus that causes ARDS. It involves symptoms that are a combination of viral pneumonia and ARDS. Antiviral or immunosuppressive medicines are used to treat many COVID-19 patients. Several drugs are now undergoing clinical studies in order to see if they can be repurposed in the future. MATERIAL AND METHODS In this study, in silico biomarker-targeted methodologies, such as target/ molecule virtual screening by docking technique and drug repositioning strategy, as well as data mining approach and meta-analysis of investigational data, were used. RESULTS In silico findings of used combination of drug repurposing and high-throughput docking methods presented acetaminophen, ursodiol, and β-carotene as a three-drug therapy regimen to treat ARDS induced by viral pneumonia in addition to inducing direct antiviral effects against COVID-19 viral infection. CONCLUSION In the current study, drug repurposing and high throughput docking methods have been employed to develop combination drug regimens as multiple-molecule drugs for the therapy of COVID-19 and ARDS based on a multiple-target therapy strategy. This approach offers a promising avenue for the treatment of COVID-19 and ARDS, and highlights the potential benefits of drug repurposing in the fight against the current pandemic.
Collapse
Affiliation(s)
- Ghazaleh Ghavami
- Drug Design and Bioinformatics Unit, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Soroush Sardari
- Drug Design and Bioinformatics Unit, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
94
|
Eftekhar Z, Haybar H, Mohebbi A, Saki N. Cardiac Complications and COVID-19: A Review of Life-threatening Co-morbidities. Curr Cardiol Rev 2024; 20:1-12. [PMID: 38415433 PMCID: PMC11284692 DOI: 10.2174/011573403x279782240206091322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/23/2023] [Accepted: 01/19/2024] [Indexed: 02/29/2024] Open
Abstract
The novel 2019 coronavirus disease (COVID-19) was first reported in the last days of December 2019 in Wuhan, China. The presence of certain co-morbidities, including cardiovascular diseases (CVDs), are the basis for worse outcomes in patients with COVID-19. Relevant English-language literature was searched and retrieved from the Google Scholar search engine and PubMed database up to 2023 using COVID-19, SARS-CoV-2, Heart failure, Myocardial infarction, and Arrhythmia and Cardiac complication as keywords. Increased hemodynamic load, ischemia-related dysfunction, ventricular remodeling, excessive neurohumoral stimulation, abnormal myocyte calcium cycling, and excessive or insufficient extracellular matrix proliferation are associated with heart failure (HF) in COVID-19 patients. Inflammatory reaction due to the excessive release of inflammatory cytokines, leads to myocardial infarction (MI) in these patients. The virus can induce heart arrhythmia through cardiac complications, hypoxia, decreased heart hemodynamics, and remarkable inflammatory markers. Moreover, studies have linked cardiac complications in COVID-19 with poor outcomes, extended hospitalization time, and increased mortality rate. Patients with COVID-19 and CVDs are at higher mortality risk and they should be given high priority when receiving the treatment and intensive care during hospitalization.
Collapse
Affiliation(s)
- Zeinab Eftekhar
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Mohebbi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
95
|
Wu MA, Del GIovane C, Colombo R, Dolci G, Arquati M, Vicini R, Russo U, Ruggiero D, Coluccio V, Taino A, Franceschini E, Facchinetti P, Mighali P, Trombetta L, Tonelli F, Gabiati C, Cogliati C, D'Amico R, Marietta M. Low-molecular-weight heparin for the prevention of clinical worsening in severe non-critically ill COVID-19 patients: a joint analysis of two randomized controlled trials. Intern Emerg Med 2024; 19:71-79. [PMID: 37794281 DOI: 10.1007/s11739-023-03439-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/14/2023] [Indexed: 10/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19) carries a high risk of vascular thrombosis. However, whether a specific anticoagulation intensity strategy may prevent clinical worsening in severe COVID-19 patients is still debated. We conducted a joint analysis of two randomized controlled trials, COVID-19 HD (NCT044082359) and EMOS-COVID (NCT04646655), to assess the efficacy and safety of two anticoagulant regimens in hospitalized severe COVID-19 patients. Subjects with COVID-19-associated respiratory compromise and/or coagulopathy were randomly assigned to low (4000 IU qd) or high (70 IU Kg-1 every 12 h) enoxaparin dose. The primary efficacy endpoint was clinical worsening within 30 days, defined as the occurrence of at least one of the following events, whichever came first: in-hospital death, evidence of arterial or venous thromboembolism, acute myocardial infarction, need for either continuous positive airway pressure (CPAP) or non-invasive ventilation (NIV) in patients receiving standard oxygen therapy or none at randomization, and need for mechanical ventilation in any patient. The safety endpoint was major bleeding. We estimated the relative risk (RR) and its 95% confidence interval (CI) for the outcomes. Among 283 patients included in the study (144 in the low-dose and 139 in the high-dose group), 118 (41.7%) were on NIV or CPAP at randomization. 23/139 (16.5%) patients in the high-dose group reached the primary endpoint compared to 33/144 (22.9%) in the low-dose group (RR 0.72, 95% CI 0.45-1.17). No major bleeding was observed. No significant differences were found in the clinical worsening of hospitalized COVID-19 patients treated with high versus low doses of enoxaparin.
Collapse
Affiliation(s)
- Maddalena Alessandra Wu
- Division of Internal Medicine, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy.
| | - Cinzia Del GIovane
- Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
- Unità di Supporto Statistico Metodologico per la Ricerca Clinica Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Riccardo Colombo
- Division of Anesthesiology and Intensive Care, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Giovanni Dolci
- Infectious Diseases Unit, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Roberto Vicini
- Unità di Supporto Statistico Metodologico per la Ricerca Clinica Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Umberto Russo
- Division of Haematology, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Diego Ruggiero
- Division of Cardiology, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Valeria Coluccio
- Hematology Unit, Azienda Ospedaliero-Universitaria, Modena, Italy
| | - Alba Taino
- Division of Internal Medicine, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Erica Franceschini
- Infectious Diseases Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Pietro Facchinetti
- Division of Internal Medicine, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Pasquale Mighali
- Servizio Formazione, Ricerca e Innovazione, Azienda Ospedaliero-Universitaria di Modena, Ospedale Policlinico, Modena, Italy
| | - Lucia Trombetta
- Division of Internal Medicine, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Francesca Tonelli
- Division of Internal Medicine, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Claudia Gabiati
- Division of Internal Medicine, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Chiara Cogliati
- Division of Internal Medicine, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Roberto D'Amico
- Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
- Unità di Supporto Statistico Metodologico per la Ricerca Clinica Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Marco Marietta
- Hematology Unit, Azienda Ospedaliero-Universitaria, Modena, Italy
| |
Collapse
|
96
|
Yong J, Toh CH. Rethinking coagulation: from enzymatic cascade and cell-based reactions to a convergent model involving innate immune activation. Blood 2023; 142:2133-2145. [PMID: 37890148 DOI: 10.1182/blood.2023021166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
ABSTRACT Advancements in the conceptual thinking of hemostasis and thrombosis have been catalyzed by major developments within health research over several decades. The cascade model of coagulation was first described in the 1960s, when biochemistry gained prominence through innovative experimentation and technical developments. This was followed by the cell-based model, which integrated cellular coordination to the enzymology of clot formation and was conceptualized during the growth period in cell biology at the turn of the millennium. Each step forward has heralded a revolution in clinical therapeutics, both in procoagulant and anticoagulant treatments to improve patient care. In current times, the COVID-19 pandemic may also prove to be a catalyst: thrombotic challenges including the mixed responses to anticoagulant treatment and the vaccine-induced immune thrombotic thrombocytopenia have exposed limitations in our preexisting concepts while simultaneously demanding novel therapeutic approaches. It is increasingly clear that innate immune activation as part of the host response to injury is not separate but integrated into adaptive clot formation. Our review summarizes current understanding of the major molecules facilitating such a cross talk between immunity, inflammation and coagulation. We demonstrate how such effects can be layered upon the cascade and cell-based models to evolve conceptual understanding of the physiology of immunohemostasis and the pathology of immunothrombosis.
Collapse
Affiliation(s)
- Jun Yong
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
97
|
Alshahrani A, Almoahzieie A, Alshareef H, Alammash BB, Alhamidi S, Meraya AM, Alshammari AS, Ajlan A, Alghofaili A, Alnassar A, Alshahrani N, Aldossari M, Alkhaldi T, Alwazzeh MJ, Almashouf AB, Alkuwaiti FA, Alghamdi SH, Alshehri O, Ali M. Death and Venous Thromboembolism Analyses among Hospitalized COVID-19-Positive Patients: A Multicenter Study. J Clin Med 2023; 12:7624. [PMID: 38137692 PMCID: PMC10743652 DOI: 10.3390/jcm12247624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/26/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Coagulation disorders are frequently encountered among patients infected with coronavirus disease 2019 (COVID-19), especially among admitted patients with more severe symptoms. This study aims to determine the mortality rate and incidence and risk factors for venous thromboembolism (VTE) in hospitalized patients with COVID-19. METHODS This retrospective observational cohort study was conducted from March to July 2020 using a hospital database. All adult patients (>18 years old) with laboratory-confirmed COVID-19 were included. Laboratory data and the real-time reverse transcriptase-polymerase chain reaction (rRT-PCR) for SARS-CoV-2 were obtained from medical records. The mortality rate and the incidence of VTE were established as study results. A multivariate logistic regression analysis was performed to identify predictors of thrombotic events. RESULTS rA total of 1024 confirmed COVID-19 patients were treated, of whom 110 (10.7%) were deceased and 58 patients (5.7%) developed VTE. Death occurred more frequently in patients older than 50 years and those admitted to the intensive care unit (ICU, 95%) and who received mechanical ventilation (62.7%). Multivariate analysis revealed that cancer patients were two times more likely to have VTE (adjusted odds ratio = 2.614; 95% CI = (1.048-6.519); p = 0.039). Other chronic diseases, such as diabetes, hypertension, and chronic kidney disease, were not associated with an increased risk of VTE. CONCLUSIONS One-tenth of hospitalized COVID-19 patients were deceased, and VTE was prevalent among patients with chronic conditions, such as cancer, despite anticoagulation therapy. Healthcare professionals should closely monitor individuals with a high risk of developing VTE to prevent unwanted complications.
Collapse
Affiliation(s)
- Asma Alshahrani
- Department of Clinical Pharmacy, Faculty of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia;
| | - Abdullah Almoahzieie
- Pharmaceutical Care Division, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia; (A.A.); (A.A.); (A.A.)
- Clinical Pharmacy Department, College of Pharmacy, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Hanan Alshareef
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk 47512, Saudi Arabia; (H.A.); (M.A.)
| | - Buthinah B. Alammash
- Department of Pharmaceutical care services, King Fahad Hospital, Ministry of Health, AL Madinah Munawara 42351, Saudi Arabia;
| | - Sarah Alhamidi
- Pharmaceutical Care Division, Security Forces Hospital, Riyadh 11481, Saudi Arabia;
| | - Abdulkarim M. Meraya
- Department of Clinical Pharmacy, Pharmacy Practice Research, College of Pharmacy, Jazan University, Jazan 82722, Saudi Arabia;
| | - Abdullah S. Alshammari
- Pharmaceutical Practice Department, College of Pharmacy, Umm Al-Qura University, Makkah 24382, Saudi Arabia;
- Department of Clinical Pharmacy, King Abdullah Medical City, Makkah 24331, Saudi Arabia
| | - Aziza Ajlan
- Pharmaceutical Care Division, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia; (A.A.); (A.A.); (A.A.)
| | - Alnajla Alghofaili
- Pharmaceutical Care Division, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia; (A.A.); (A.A.); (A.A.)
| | - Abdullah Alnassar
- Pharmaceutical Care Division, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia; (A.A.); (A.A.); (A.A.)
| | - Nada Alshahrani
- Department of Internal Medicine, Prince Sultan Medical City, Riyadh 12624, Saudi Arabia;
| | - Maram Aldossari
- Pharmaceutical Care Division, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
| | - Turkiah Alkhaldi
- Department of Clinical Pharmacy, King Fahad Medical City, Riyadh 11525, Saudi Arabia
| | - Marwan J. Alwazzeh
- Department of Internal Medicine, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Abdullah B. Almashouf
- Department of Internal Medicine, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Feras A. Alkuwaiti
- Department of Internal Medicine, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Shrouq Hamed Alghamdi
- Department of Pharmacy, Prince Muhammad bin Abdualaziz Hospital, Ministry of Health, Riyadh 12769, Saudi Arabia
| | - Ohuod Alshehri
- Department of Clinical Pharmacy, Faculty of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia;
| | - Mostafa Ali
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk 47512, Saudi Arabia; (H.A.); (M.A.)
- Department of Clinical Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| |
Collapse
|
98
|
Córneo ES, Veras FP, Gomes GF, Schneider AH, Manuella B, Almeida CJLR, Silva CM, Martins RB, Batah SS, Simon CS, Prestes GDS, Alves-Filho JC, Arruda E, Louzada-Junior P, de Oliveira RDR, Fabro AT, Cunha TM, Cunha FQ, Dal-Pizzol F. Enoxaparin improves COVID-19 by reducing Neutrophils Extracellular Traps (NETs) production. Clin Immunol 2023; 257:109836. [PMID: 37951516 DOI: 10.1016/j.clim.2023.109836] [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: 08/03/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND COVID-19 causes consequences such as imbalance of the immune system and thrombotic events. During the infection process, NETs in excess induce a pro-inflammatory response and disseminated intravascular coagulation. We evaluated the role of enoxaparin as a potential inhibitor of NETs. METHODS K18-hACE2 animals infected with the SARS-CoV-2 virus and a group of 23 individuals admitted to the hospital with COVID-19 treated with enoxaparin or without treatment and controls without the disease were included. RESULTS Enoxaparin decreased the levels of NETs, reduced the signs of the disease and mitigated lung damage in the animals infected with SARS-CoV-2. These effects were partially associated with prevention of SARS-CoV-2 entry and NETs synthesis. Clinical data revealed that treatment with enoxaparin decreased the levels of inflammatory markers, the levels of NETs in isolated neutrophils and the organ dysfunction. CONCLUSION This study provides evidence for the beneficial effects of enoxaparin in COVID-19 in addition to its anticoagulant role.
Collapse
Affiliation(s)
- Emily S Córneo
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil.
| | - Flavio Protasio Veras
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil; Departments of Pharmacology Research Center; Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Giovanni F Gomes
- Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil; Departments of Pharmacology Research Center
| | - Ayda H Schneider
- Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil; Departments of Pharmacology Research Center
| | - Bruna Manuella
- Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil; Departments of Pharmacology Research Center
| | - Cicero J L R Almeida
- Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil; Departments of Pharmacology Research Center
| | - Camila M Silva
- Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil; Departments of Pharmacology Research Center
| | | | - Sabrina S Batah
- Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carla S Simon
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Gabriele da S Prestes
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - José Carlos Alves-Filho
- Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil; Departments of Pharmacology Research Center
| | | | - Paulo Louzada-Junior
- Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil; Internal Medicine, Division of Clinical Immunology and Division of Infectious Diseases
| | - Renê D R de Oliveira
- Internal Medicine, Division of Clinical Immunology and Division of Infectious Diseases
| | - Alexandre T Fabro
- Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thiago M Cunha
- Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil; Departments of Pharmacology Research Center
| | - Fernando Queiroz Cunha
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil; Departments of Pharmacology Research Center
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| |
Collapse
|
99
|
Liu L, Zhou C, Jiang H, Wei H, Zhou Y, Zhou C, Ji X. Epidemiology, pathogenesis, and management of Coronavirus disease 2019-associated stroke. Front Med 2023; 17:1047-1067. [PMID: 38165535 DOI: 10.1007/s11684-023-1041-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/15/2023] [Indexed: 01/03/2024]
Abstract
The Coronavirus disease 2019 (COVID-19) epidemic has triggered a huge impact on healthcare, socioeconomics, and other aspects of the world over the past three years. An increasing number of studies have identified a complex relationship between COVID-19 and stroke, although active measures are being implemented to prevent disease transmission. Severe COVID-19 may be associated with an increased risk of stroke and increase the rates of disability and mortality, posing a serious challenge to acute stroke diagnosis, treatment, and care. This review aims to provide an update on the influence of COVID-19 itself or vaccines on stroke, including arterial stroke (ischemic stroke and hemorrhagic stroke) and venous stroke (cerebral venous thrombosis). Additionally, the neurovascular mechanisms involved in SARS-CoV-2 infection and the clinical characteristics of stroke in the COVID-19 setting are presented. Evidence on vaccinations, potential therapeutic approaches, and effective strategies for stroke management has been highlighted.
Collapse
Affiliation(s)
- Lu Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
| | - Chenxia Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
| | - Huimin Jiang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Huimin Wei
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yifan Zhou
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Chen Zhou
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China.
- Department of Neurology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| | - Xunming Ji
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China.
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China.
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China.
| |
Collapse
|
100
|
Wang T, Zhao Z, Li W, Wu J, Ye Q, Xie H. Machine Learning Predictive Modeling for the Identification of Moderate Coronavirus Disease 2019 During the Pandemic: A Retrospective Study. Cureus 2023; 15:e50619. [PMID: 38226092 PMCID: PMC10789081 DOI: 10.7759/cureus.50619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2023] [Indexed: 01/17/2024] Open
Abstract
BACKGROUND Timely differentiation of moderate COVID-19 cases from mild cases is beneficial for early treatment and saves medical resources during the pandemic. We attempted to construct a model to predict the occurrence of moderate COVID-19 through a retrospective study. METHODS In this retrospective study, clinical data from patients with COVID-19 admitted to Hainan Western Central Hospital in Danzhou, China, between August 1, 2022, and August 31, 2022, was collected, including sex, age, signs on admission, comorbidities, imaging data, post-admission treatment, length of stay, and the results of laboratory tests on admission. The patients were classified into a mild-to-moderate-type group according to WHO guidance. Factors that differed between groups were included in machine learning models such as Bernoulli Naïve Bayes (BNB), linear discriminant analysis, support vector machine (SVM), least absolute shrinkage and selection operator (LASSO), and logistic regression (LR) models. These models were compared to select the optimal model with the best predictive efficacy for moderate COVID-19. The predictive performance of the models was assessed using the area under the curve (AUC), sensitivity, specificity, and calibration plot. RESULTS A total of 231 patients with COVID-19 were included in this retrospective analysis. Among them, 152 (68.83%) were mild types, 72 (31.17%) were moderate types, and there were no patients with severe or critical types. A logistic regression model combined with age, respiratory rate (RR), lactate dehydrogenase (LDH), D-dimer, and albumin was selected to predict the occurrence of moderate COVID-19. The receiver operating characteristic curve (ROC) showed that AUC, sensitivity, and specificity in the model were 0.719, 0.681, and 0.635, respectively, in predicting moderate COVID-19. Calibration curve analysis revealed that the predicted probability of the model was in good agreement with the true probability. Stratified analysis showed better predictive efficacy after modeling for people aged ≤66 years (AUC = 0.7656) and a better calibration curve. CONCLUSION The LR model, combined with age, RR, D-dimer, LDH, and albumin, can predict the occurrence of moderate COVID-19 well, especially for patients aged ≤66 years.
Collapse
Affiliation(s)
- Tao Wang
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai, CHN
| | - Zhanqing Zhao
- Department of Critical Care Medicine, Hainan Western Central Hospital, Danzhou, CHN
| | - Wenzhe Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, CHN
| | - Jing Wu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, CHN
| | - Qianru Ye
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, CHN
| | - Hui Xie
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai, CHN
| |
Collapse
|