1
|
Taniguchi H, Abe T, Takeuchi I, Ohshimo S, Shime N, Kushimoto S, Hashimoto S, Takeda S. Characteristics of Bleeding Complications in Patients with Severe COVID-19 Requiring Veno-venous Extracorporeal Membrane Oxygenation in Japan. Thromb Haemost 2024. [PMID: 39242097 DOI: 10.1055/a-2411-1000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
BACKGROUND Complications during veno-venous extracorporeal membrane oxygenation (VV-ECMO) are associated with in-hospital mortality. Asian patients on extracorporeal membrane oxygenation (ECMO) have higher risks of bleeding and in-hospital mortality than Caucasian patients. This study aimed to characterize and identify bleeding complications and their associated factors related to in-hospital mortality in patients with severe coronavirus disease 2019 (COVID-19) requiring VV-ECMO in Japan. METHODS In this retrospective observational analysis, the prospective nationwide multicenter registry was used to track real-time information from intensive care units throughout Japan during the COVID-19 pandemic. VV-ECMO patients' registry data between February 1, 2020 and October 31, 2022 were used. RESULTS This study included 441 patients; 178 (40%) had bleeding complications in the following sites: 20% at the cannulation site, 16% in the gastrointestinal tract, 16% in the ear-nose-throat, 13% at the tracheostomy site, 9% intrathoracic, 6% intracranial, and 5% in the iliopsoas. Anticoagulation was discontinued in >50% of patients with intracranial, iliopsoas, and gastrointestinal tract bleeding. ECMO was discontinued in one-third of patients with intracranial, intramuscular, and iliopsoas hemorrhages. Multivariable logistic regression analysis revealed that only gastrointestinal tract bleeding was associated with in-hospital mortality (odds ratio: 2.49; 95% confidence interval: 1.11-5.60; p = 0.03). CONCLUSION Incidence of bleeding complications was 40% in the Japanese population. Gastrointestinal tract bleeding emerged as a significant predictor of adverse outcomes, necessitating further research into preventive strategies and optimized care protocols. These findings can guide the management of VV-ECMO patients with COVID-19.
Collapse
Affiliation(s)
- Hayato Taniguchi
- Department of Emergency Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Non-profit Organization Japan ECMO Network, Tokyo, Japan
| | - Takeru Abe
- Department of Emergency Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ichiro Takeuchi
- Department of Emergency Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Non-profit Organization Japan ECMO Network, Tokyo, Japan
| | - Shinichiro Ohshimo
- Non-profit Organization Japan ECMO Network, Tokyo, Japan
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobuaki Shime
- Non-profit Organization Japan ECMO Network, Tokyo, Japan
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shigeki Kushimoto
- Non-profit Organization Japan ECMO Network, Tokyo, Japan
- Division of Emergency and Critical Care Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoru Hashimoto
- Non-profit Organization Japan ECMO Network, Tokyo, Japan
- Non-Profit Organization ICU Collaboration Network (ICON), Tokyo, Japan
| | - Shinhiro Takeda
- Non-profit Organization Japan ECMO Network, Tokyo, Japan
- Kawaguchi Cardiovascular and Respiratory Hospital, Kawaguchi, Japan
| |
Collapse
|
2
|
Chen Y, Li Y, Zhao Y, Pei L, Zhang L, Zou D. NanoLuciferase technology-based detection of TMPRSS2 as attempt to develop anti-coronavirus agents. Biochem Biophys Rep 2024; 39:101783. [PMID: 39156721 PMCID: PMC11326902 DOI: 10.1016/j.bbrep.2024.101783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 08/20/2024] Open
Abstract
•Utilized NanoBiT technology for high-throughput screening.•Identified compounds reducing TMPRSS2 expression, a crucial step for SARS-CoV-2 entry.•Explored a broad range of compound libraries.
Collapse
Affiliation(s)
- Yanwen Chen
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai, 200025, China
| | - Yunqi Li
- National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai, 200025, China
| | - Ye Zhao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai, 200025, China
| | - Lei Pei
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai, 200025, China
| | - Ling Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai, 200025, China
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai, 200025, China
| |
Collapse
|
3
|
Lou Y, Li Z, Zheng H, Yuan Z, Li W, Zhang J, Shen W, Gao Y, Ran N, Kong X, Feng S. New strategy to treat spinal cord injury: Nafamostat mesilate suppressed NLRP3-mediated pyroptosis during acute phase. Int Immunopharmacol 2024; 134:112190. [PMID: 38703569 DOI: 10.1016/j.intimp.2024.112190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024]
Abstract
Spinal cord injury (SCI) is a devastating condition for which effective clinical treatment is currently lacking. During the acute phase of SCI, myriad pathological changes give rise to subsequent secondary injury. The results of our previous studies indicated that treating rats post-SCI with nafamostat mesilate (NM) protected the blood-spinal cord barrier (BSCB) and exerted an antiapoptotic effect. However, the optimal dosage for mice with SCI and the underlying mechanisms potentially contributing to recovery, especially during the acute phase of SCI, have not been determined. In this study, we first determined the optimal dosage of NM for mice post-SCI (5 mg/kg/day). Subsequently, our RNA-seq findings revealed that NM has the potential to inhibit pyroptosis after SCI. These findings were further substantiated by subsequent Western blot (WB) and Immunofluorescence (IF) analyses in vivo. These results indicate that NM can alleviate NLRP3 (NOD-like receptor thermal protein domain associated protein 3)-mediated pyroptosis by modulating the NF-κB signaling pathway and reducing the protein expression levels of NIMA-related kinase 7 (NEK7) and cathepsin B (CTSB). In vitro experimental results supported our in vivo findings, revealing the effectiveness of NM in suppressing pyroptosis induced by adenosine triphosphate (ATP) and lipopolysaccharide (LPS) in BV2 cells. These results underscore the potential of NM to regulate NLRP3-mediated pyroptosis following SCI. Notably, compared with other synthetic compounds, NM exhibits greater versatility, suggesting that it is a promising clinical treatment option for SCI.
Collapse
Affiliation(s)
- Yongfu Lou
- Department of Orthopedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Zonghao Li
- Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China
| | - Han Zheng
- Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China
| | - Zhongze Yuan
- Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China
| | - Wenxiang Li
- Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China
| | - Jianping Zhang
- Division of Surgery and Interventional Science, University College London, London HA7 4LP, United Kingdom
| | - Wenyuan Shen
- Department of Orthopedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Yiming Gao
- Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China
| | - Ning Ran
- Department of Orthopedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China.
| | - Xiaohong Kong
- Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China.
| | - Shiqing Feng
- Department of Orthopedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China.
| |
Collapse
|
4
|
Wei M, Li T, Liu S, Wang Y, Tran C, Ao G. The Effectiveness and Safety of Nafamostat Mesylate in the Treatment of COVID-19: a Meta-Analysis. Jpn J Infect Dis 2024; 77:182-186. [PMID: 38296543 DOI: 10.7883/yoken.jjid.2023.315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
Nafamostat mesylate, a synthetic serine protease inhibitor, has been shown to have antiviral activity against SARS-CoV-2 and anticoagulant properties that may be beneficial in the treatment of COVID-19. We conducted a meta-analysis to evaluate the effectiveness and safety of nafamostat mesylate for the treatment of COVID-19. PubMed, Embase, Cochrane Library, Scopus, Web of Science, medRxiv, and bioRxiv were searched up to July 2023 for studies comparing the outcomes of nafamostat mesylate treatment and no nafamostat mesylate treatment in patients with COVID-19. Mortality, disease progression, and adverse events were analyzed. Six studies involving 16,195 patients were included in the analysis. Meta-analysis revealed no significant difference in mortality (odds ratio [OR]: 0.88, 95% CI: 0.20-3.75, P = 0.86) or disease progression (OR: 2.76, 95% CI: 0.31-24.68, P = 0.36) between groups. However, nafamostat mesylate was associated with an increased risk of hyperkalemia (OR: 7.15, 95% CI: 2.66-19.24, P < 0.0001). Nafamostat mesylate did not improve mortality or morbidity in hospitalized patients with COVID-19. The risk of hyperkalemia is a serious concern that requires monitoring and preventive measures. Further research in different COVID-19 populations is required.
Collapse
Affiliation(s)
- Mian Wei
- Department of Nephrology, Chengdu First People's Hospital, China
| | - Toni Li
- Division of Emergency Medicine, Department of Medicine, University of Toronto, Canada
| | - Siyuan Liu
- Key laboratory of Shanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, China
| | - Yushu Wang
- Chengdu West China Clinical Research Center, China
| | - Carolyn Tran
- Schulich School of Medicine & Dentistry, Western University, Canada
| | - Guangyu Ao
- Department of Nephrology, Chengdu First People's Hospital, China
| |
Collapse
|
5
|
Li P, Kim Y, Dampalla CS, Nhat Nguyen H, Meyerholz DK, Johnson DK, Lovell S, Groutas WC, Perlman S, Chang KO. Potent 3CLpro inhibitors effective against SARS-CoV-2 and MERS-CoV in animal models by therapeutic treatment. mBio 2024; 15:e0287823. [PMID: 38126789 PMCID: PMC10865860 DOI: 10.1128/mbio.02878-23] [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: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 12/23/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Middle East respiratory syndrome coronavirus (MERS-CoV) are zoonotic betacoronaviruses that continue to have a significant impact on public health. Timely development and introduction of vaccines and antivirals against SARS-CoV-2 into the clinic have substantially mitigated the burden of COVID-19. However, a limited or lacking therapeutic arsenal for SARS-CoV-2 and MERS-CoV infections, respectively, calls for an expanded and diversified portfolio of antivirals against these coronavirus infections. In this report, we examined the efficacy of two potent 3CLpro inhibitors, 5d and 11d, in fatal animal models of SARS-CoV-2 and MERS-CoV to demonstrate their broad-spectrum activity against both viral infections. These compounds significantly increased the survival of mice in both models when treatment started 1 day post infection compared to no treatment which led to 100% fatality. Especially, the treatment with compound 11d resulted in 80% and 90% survival in SARS-CoV-2 and MERS-CoV-infected mice, respectively. Amelioration of lung viral load and histopathological changes in treated mice correlated well with improved survival in both infection models. Furthermore, compound 11d exhibited significant antiviral activities in K18-hACE2 mice infected with SARS-CoV-2 Omicron subvariant XBB.1.16. The results suggest that these are promising candidates for further development as broad-spectrum direct-acting antivirals against highly virulent human coronaviruses.IMPORTANCEHuman coronaviruses such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Middle East respiratory syndrome coronavirus (MERS-CoV) continue to have a significant impact on public health. A limited or lacking therapeutic arsenal for SARS-CoV-2 and MERS-CoV infections calls for an expanded and diversified portfolio of antivirals against these coronavirus infections. We have previously reported a series of small-molecule 3C-like protease (3CLpro) inhibitors against human coronaviruses. In this report, we demonstrated the in vivo efficacy of 3CLpro inhibitors for their broad-spectrum activity against both SARS-CoV-2 and MERS-CoV infections using the fatal animal models. The results suggest that these are promising candidates for further development as broad-spectrum direct-acting antivirals against highly virulent human coronaviruses.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Microbiology and Immunology, The University of Iowa, lowa, USA
| | - Yunjeong Kim
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | | | - Harry Nhat Nguyen
- Department of Chemistry, Wichita State University, Wichita, Kansas, USA
| | | | - David K. Johnson
- Computational Chemical Biology Core, The University of Kansas, Lawrence, Kansas, USA
| | - Scott Lovell
- Protein Structure Laboratory, The University of Kansas, Lawrence, Kansas, USA
| | | | - Stanley Perlman
- Department of Microbiology and Immunology, The University of Iowa, lowa, USA
| | - Kyeong-Ok Chang
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
6
|
Liu K, Li ZH. Efficacy and safety of Nafamostat mesylate in patients with end-stage renal failure. World J Clin Cases 2024; 12:68-75. [PMID: 38292627 PMCID: PMC10824189 DOI: 10.12998/wjcc.v12.i1.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/05/2023] [Accepted: 12/18/2023] [Indexed: 01/02/2024] Open
Abstract
BACKGROUND Recent studies on dialysis anticoagulation therapy in patients with renal failure have shown that Nafamostat mesylate, a broad-spectrum potent serine protease inhibitor, has strong anticoagulation and anti-fiber activity. AIM To evaluate the efficacy and safety of Nafamostat mesylate in patients with end-stage renal failure. METHODS Seventy-five patients with end-stage renal failure who received hemodialysis at our hospital between January 2020 and August 2021 were selected and divided into the observation group (Nafamostat mesylate for injection, n = 33) and control group (heparin sodium injection, n = 32). General patient data, indicators of clinical efficacy, dialyzer hemocoagulation parameters, coagulation function indices, and hemoglobin concentration and platelet count before and after treatment, and the occurrence of adverse reactions after treatment were compared between the two groups. RESULTS The two groups showed no significant differences in general patient data (P > 0.05). The post-treatment effectiveness rate in the control group was lower than that in the observation group (P < 0.05). The two groups showed no significant difference in the number of patients in grade I (P > 0.05), while the number of patients in grade 0 was lower in the control group, and the number of patients in grades II and III was higher in the control group (P < 0.05). The post-treatment prothrombin time, activated partial thromboplastin time, thrombin time, and international normalized ratio values in the control group were higher than those in the observation group, while the fibrinogen level in the control group was lower than that in the observation group (P < 0.05). The two groups showed no significant difference in the platelet count and hemoglobin level before and after treatment (P > 0.05). The total number of post-treatment adverse reactions in the observation group was lower than that in the control group (P < 0.05). CONCLUSION Treatment of patients showing end-stage renal failure with Nafamostat mesylate can significantly improve therapeutic efficacy and has high safety and clinical value.
Collapse
Affiliation(s)
- Kun Liu
- Department of Critical Care Medicine, The Third People's Hospital Of Hubei Province, Wuhan 430030, Hubei Province, China
| | - Zhen-Hua Li
- Department of Critical Care Medicine, General Hospital of The Yangtze River Shipping, Wuhan Brain Hospital, Wuhan 430015, Hubei Province, China
| |
Collapse
|
7
|
Narumi K, Okada T, Lin Y, Kikuchi S. Efficacy of nafamostat mesylate in the prevention of pancreatitis after endoscopic retrograde cholangiopancreatography: a systematic review and meta-analysis of randomized controlled trials. Sci Rep 2023; 13:23012. [PMID: 38155200 PMCID: PMC10754829 DOI: 10.1038/s41598-023-50181-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023] Open
Abstract
We conducted a systematic review and meta-analysis to evaluate the effect of nafamostat on the prevention of post-endoscopic retrograde cholangiopancreatography (ERCP) pancreatitis (PEP). PubMed, Web of Science, and Ichushi Web were searched for randomized controlled trials (RCTs) using nafamostat to prevent PEP. In subgroup analyses, we studied the preventive effects of nafamostat according to the severity of PEP, risk category, and dose. A random-effects model was adopted; heterogeneity between studies was examined using the chi-squared test and I2 statistics. This analysis uses the PRISMA statement as general guidance. 9 RCTs involving 3321 patients were included. The risk of PEP was lower in the nafamostat group than in the control group [4.4% vs. 8.3%, risk ratio (RR): 0.50, 95% confidence interval (CI): 0.36-0.68]. In subgroup analyses, the protective effects were evident in low-risk patients for PEP before ERCP (RR: 0.34, 95% CI: 0.21-0.55). The association between PEP and nafamostat was significant only in patients who developed mild PEP (RR: 0.49; 95% CI: 0.36-0.69). The benefits were independent of the dose. The prophylactic use of nafamostat resulted in a lower risk of PEP. The subgroup analyses suggested uncertain benefits for severe PEP or high-risk patients for PEP. This warrants further investigation through additional RCTs.
Collapse
Affiliation(s)
- Kazuaki Narumi
- Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Tomoki Okada
- Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Yingsong Lin
- Department of Public Health, Aichi Medical University School of Medicine, Yazakokarimata 1-1, Nagakute, Aichi, 480-1195, Japan.
| | - Shogo Kikuchi
- Department of Public Health, Aichi Medical University School of Medicine, Yazakokarimata 1-1, Nagakute, Aichi, 480-1195, Japan
| |
Collapse
|
8
|
Tang S, Xu L, Li H, Wu Z, Wen Q. Anticoagulants in adult extracorporeal membrane oxygenation: alternatives to standardized anticoagulation with unfractionated heparin. Eur J Clin Pharmacol 2023; 79:1583-1594. [PMID: 37740749 DOI: 10.1007/s00228-023-03568-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/14/2023] [Indexed: 09/25/2023]
Abstract
BACKGROUND Extracorporeal membrane oxygenation (ECMO) is a vital technique for severe respiratory or heart failure patients. Bleeding and thrombotic events are common during ECMO and negatively impact patient outcomes. Unfractionated heparin is the primary anticoagulant, but its adverse effects limit its use, necessitating alternative anticoagulants. OBJECTIVE Review available alternative anticoagulants for adult ECMO patients. Explore potential novel anticoagulants for future ECMO use. Aim to reduce complications (bleeding and thrombosis) and improve safety and efficacy for critically ill ECMO patients. METHODS Comprehensive literature review of existing and emerging anticoagulants for ECMO. RESULTS Identified a range of alternative anticoagulants beyond unfractionated heparin. Evaluated their potential utility in mitigating ECMO-related complications. CONCLUSION Diverse anticoagulant options are available and under investigation for ECMO. These alternatives may enhance patient safety and outcomes during ECMO support. Further research and clinical studies are warranted to determine their effectiveness and safety profiles.
Collapse
Affiliation(s)
- Shu Tang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Liqing Xu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhanshen Wu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiang Wen
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Dasgupta A, Gangai S, Narayan R, Kapoor S. Mapping the Lipid Signatures in COVID-19 Infection: Diagnostic and Therapeutic Solutions. J Med Chem 2023; 66:14411-14433. [PMID: 37899546 DOI: 10.1021/acs.jmedchem.3c01238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
The COVID-19 pandemic ignited research centered around the identification of robust biomarkers and therapeutic targets. SARS-CoV-2, the virus responsible, hijacks the metabolic machinery of the host cells. It relies on lipids and lipoproteins of host cells for entry, trafficking, immune evasion, viral replication, and exocytosis. The infection causes host cell lipid metabolic remodelling. Targeting lipid-based processes is thus a promising strategy for countering COVID-19. Here, we review the role of lipids in the different steps of the SARS-CoV-2 pathogenesis and identify lipid-centric targetable avenues. We discuss lipidome changes in infected patients and their relevance as potential clinical diagnostic or prognostic biomarkers. We summarize the emerging direct and indirect therapeutic approaches for targeting COVID-19 using lipid-inspired approaches. Given that viral protein-targeted therapies may become less effective due to mutations in emerging SARS-CoV-2 variants, lipid-inspired interventions may provide additional and perhaps better means of combating this and future pandemics.
Collapse
Affiliation(s)
- Aishi Dasgupta
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
- IIT-Bombay Monash Academy, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Shon Gangai
- School of Chemical and Materials Sciences (SCMS), Institute of Technology Goa, Farmagudi, Ponda, Goa 403401, India
| | - Rishikesh Narayan
- School of Chemical and Materials Sciences (SCMS), Institute of Technology Goa, Farmagudi, Ponda, Goa 403401, India
- School of Interdisciplinary Life Sciences (SILS), Institute of Technology Goa, Farmagudi, Ponda, Goa 403401, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
- IIT-Bombay Monash Academy, Indian Institute of Technology Bombay, Mumbai 400076, India
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| |
Collapse
|
10
|
Awad AM, Hansen K, Del Rio D, Flores D, Barghash RF, Kakkola L, Julkunen I, Awad K. Insights into COVID-19: Perspectives on Drug Remedies and Host Cell Responses. Biomolecules 2023; 13:1452. [PMID: 37892134 PMCID: PMC10604481 DOI: 10.3390/biom13101452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
In light of the COVID-19 global pandemic caused by SARS-CoV-2, ongoing research has centered on minimizing viral spread either by stopping viral entry or inhibiting viral replication. Repurposing antiviral drugs, typically nucleoside analogs, has proven successful at inhibiting virus replication. This review summarizes current information regarding coronavirus classification and characterization and presents the broad clinical consequences of SARS-CoV-2 activation of the angiotensin-converting enzyme 2 (ACE2) receptor expressed in different human cell types. It provides publicly available knowledge on the chemical nature of proposed therapeutics and their target biomolecules to assist in the identification of potentially new drugs for the treatment of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ahmed M. Awad
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Kamryn Hansen
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Diana Del Rio
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Derek Flores
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Reham F. Barghash
- Institute of Chemical Industries Research, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Laura Kakkola
- Institute of Biomedicine, Faculty of Medicine, University of Turku, 20014 Turku, Finland
| | - Ilkka Julkunen
- Institute of Biomedicine, Faculty of Medicine, University of Turku, 20014 Turku, Finland
- Clinical Microbiology, Turku University Hospital, 20521 Turku, Finland
| | - Kareem Awad
- Institute of Biomedicine, Faculty of Medicine, University of Turku, 20014 Turku, Finland
- Department of Therapeutic Chemistry, Institute of Pharmaceutical and Drug Industries Research, National Research Center, Dokki, Cairo 12622, Egypt
| |
Collapse
|
11
|
Okugawa S, Ikeda M, Kashiwabara K, Moritoyo T, Kohsaka T, Shimizu T, Hagiya H, Hasegawa K, Otsuka F, Miwa A, Kisimoto N, Mizoguchi A, Imamura A, Ikeuchi K, Tsutsumi T, Jubishi D, Hashimoto H, Okamoto K, Harada S, Inoue JI, Seto Y, Moriya K. Antiviral effect and safety of nafamostat mesilate in patients with mild early-onset COVID-19: An exploratory multicentre randomized controlled clinical trial. Int J Antimicrob Agents 2023; 62:106922. [PMID: 37429450 DOI: 10.1016/j.ijantimicag.2023.106922] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/03/2023] [Accepted: 07/05/2023] [Indexed: 07/12/2023]
Abstract
OBJECTIVES This study aimed to evaluate the antiviral effects and safety of nafamostat in early-onset patients with coronavirus disease 2019 (COVID-19). METHODS In this exploratory multicentre randomized controlled trial, patients were assigned to three groups within 5 days of symptom onset, with 10 participants in each group: nafamostat at either 0.2 mg/kg/h or 0.1 mg/kg/h or a standard-of-care group. The primary endpoint was area under the curve for decrease in SARS-CoV-2 viral load in nasopharyngeal samples from baseline to day 6. RESULTS Of the 30 randomized patients, 19 received nafamostat. Overall, 10 patients received low-dose nafamostat, 9 patients received high-dose nafamostat, and 10 received standard-of-care. The detected viruses were Omicron strains. The regression coefficient for area under the curve for decrease in viral load as the response variable and nafamostat dose per body weight as the explanatory variable showed a significant relationship of -40.1 (95% confidence interval, -74.1 to -6.2; P = 0.022). Serious adverse events were not observed in either group. Phlebitis occurred in ca. 50% of patients treated with nafamostat. CONCLUSIONS Nafamostat exerts virus load-reducing effects in patients with early-onset COVID-19.
Collapse
Affiliation(s)
- Shu Okugawa
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan
| | - Mahoko Ikeda
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan; Department of Infection Control and Prevention, The University of Tokyo Hospital, Tokyo, Japan
| | - Kosuke Kashiwabara
- Clinical Research Promotion Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Takashi Moritoyo
- Clinical Research Promotion Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Takao Kohsaka
- Department of Surgery, Horinouchi Hospital, Saitama, Japan
| | - Toshio Shimizu
- Department of Surgery, Horinouchi Hospital, Saitama, Japan
| | - Hideharu Hagiya
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kou Hasegawa
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Fumio Otsuka
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ayumi Miwa
- Pharmaceutical Department, Takamatsu Municipal Hospital, Kagawa, Japan
| | - Nobuhito Kisimoto
- Department of Pulmonary Medicine, Takamatsu Municipal Hospital, Kagawa, Japan
| | - Ayako Mizoguchi
- Pharmaceutical Department, Moriya Keiyu Hospital, Ibaraki, Japan
| | - Akira Imamura
- Department of Internal medicine, Moriya Keiyu Hospital, Ibaraki, Japan
| | - Kazuhiko Ikeuchi
- Division of Infectious Diseases, Advanced Clinical Research Center, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takeya Tsutsumi
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan; Division of Infectious Diseases, Advanced Clinical Research Center, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Daisuke Jubishi
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan
| | - Hideki Hashimoto
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan
| | - Koh Okamoto
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan
| | - Sohei Harada
- Department of Infection Control and Prevention, The University of Tokyo Hospital, Tokyo, Japan
| | - Jun-Ichiro Inoue
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - Yasuyuki Seto
- Department of Stomach and Esophageal Surgery, The University of Tokyo Hospital, Tokyo, Japan
| | - Kyoji Moriya
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan; Department of Infection Control and Prevention, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
12
|
Ivachtchenko AV, Ivashchenko AA, Shkil DO, Ivashchenko IA. Aprotinin-Drug against Respiratory Diseases. Int J Mol Sci 2023; 24:11173. [PMID: 37446350 PMCID: PMC10342444 DOI: 10.3390/ijms241311173] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Aprotinin (APR) was discovered in 1930. APR is an effective pan-protease inhibitor, a typical "magic shotgun". Until 2007, APR was widely used as an antithrombotic and anti-inflammatory drug in cardiac and noncardiac surgeries for reduction of bleeding and thus limiting the need for blood transfusion. The ability of APR to inhibit proteolytic activation of some viruses leads to its use as an antiviral drug for the prevention and treatment of acute respiratory virus infections. However, due to incompetent interpretation of several clinical trials followed by incredible controversy in the literature, the usage of APR was nearly stopped for a decade worldwide. In 2015-2020, after re-analysis of these clinical trials' data the restrictions in APR usage were lifted worldwide. This review discusses antiviral mechanisms of APR action and summarizes current knowledge and prospective regarding the use of APR treatment for diseases caused by RNA-containing viruses, including influenza and SARS-CoV-2 viruses, or as a part of combination antiviral treatment.
Collapse
Affiliation(s)
- Alexandre V. Ivachtchenko
- ChemDiv Inc., San Diego, CA 92130, USA; (A.A.I.); (I.A.I.)
- ASAVI LLC, 1835 East Hallandale Blvd #442, Hallandale Beach, FL 33009, USA;
| | | | - Dmitrii O. Shkil
- ASAVI LLC, 1835 East Hallandale Blvd #442, Hallandale Beach, FL 33009, USA;
| | | |
Collapse
|
13
|
Hernández-Mitre MP, Won H, Wallis SC, Parker SL, Roberts JA. Stability of nafamostat in intravenous infusion solutions, human whole blood and extracted plasma: implications for clinical effectiveness studies. Bioanalysis 2023; 15:673-681. [PMID: 37272603 DOI: 10.4155/bio-2023-0040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023] Open
Abstract
Aim: To describe the stability of nafamostat in infusion solutions, during blood sample collection and in extracted plasma samples in the autosampler. Methods: Nafamostat infusion solutions were stored at room temperature in the light for 24 h. For sample collection stability, fresh blood spiked with nafamostat was subjected to combinations of anticoagulants, added esterase inhibitor and temperature. Nafamostat was monitored in the extracted plasma samples in the autosampler. Results: Nafamostat was stable in infusion solutions. Nafamostat in whole blood was stable for 3 h before centrifugation when collected in sodium fluoride/potassium oxalate tubes (4°C). Nafamostat in extracted plasma samples degraded at 4.7 ± 0.7% per h. Conclusion: Viable samples can be obtained using blood collection tubes with sodium fluoride, chilling and processing promptly.
Collapse
Affiliation(s)
| | - Hayoung Won
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, 4029, Australia
| | - Steven C Wallis
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, 4029, Australia
| | - Suzanne L Parker
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, 4029, Australia
| | - Jason A Roberts
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, 4029, Australia
- Herston Infectious Diseases Institute (HeIDI), Metro North Health, Brisbane, 4029, Australia
- Departments of Pharmacy & Intensive Care Medicine, Royal Brisbane & Women's Hospital, Brisbane, 4029, Australia
- Division of Anaesthesiology Critical Care Emergency & Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, 30029, France
| |
Collapse
|
14
|
Zabiegala A, Kim Y, Chang KO. Roles of host proteases in the entry of SARS-CoV-2. ANIMAL DISEASES 2023; 3:12. [PMID: 37128508 PMCID: PMC10125864 DOI: 10.1186/s44149-023-00075-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/07/2023] [Indexed: 05/03/2023] Open
Abstract
The spike protein (S) of SARS-CoV-2 is responsible for viral attachment and entry, thus a major factor for host susceptibility, tissue tropism, virulence and pathogenicity. The S is divided with S1 and S2 region, and the S1 contains the receptor-binding domain (RBD), while the S2 contains the hydrophobic fusion domain for the entry into the host cell. Numerous host proteases have been implicated in the activation of SARS-CoV-2 S through various cleavage sites. In this article, we review host proteases including furin, trypsin, transmembrane protease serine 2 (TMPRSS2) and cathepsins in the activation of SARS-CoV-2 S. Many betacoronaviruses including SARS-CoV-2 have polybasic residues at the S1/S2 site which is subjected to the cleavage by furin. The S1/S2 cleavage facilitates more assessable RBD to the receptor ACE2, and the binding triggers further conformational changes and exposure of the S2' site to proteases such as type II transmembrane serine proteases (TTPRs) including TMPRSS2. In the presence of TMPRSS2 on the target cells, SARS-CoV-2 can utilize a direct entry route by fusion of the viral envelope to the cellular membrane. In the absence of TMPRSS2, SARS-CoV-2 enter target cells via endosomes where multiple cathepsins cleave the S for the successful entry. Additional host proteases involved in the cleavage of the S were discussed. This article also includes roles of 3C-like protease inhibitors which have inhibitory activity against cathepsin L in the entry of SARS-CoV-2, and discussed the dual roles of such inhibitors in virus replication.
Collapse
Affiliation(s)
- Alexandria Zabiegala
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, 1800 Denison Avenue, Manhattan, KS 66506 USA
| | - Yunjeong Kim
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, 1800 Denison Avenue, Manhattan, KS 66506 USA
| | - Kyeong-Ok Chang
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, 1800 Denison Avenue, Manhattan, KS 66506 USA
| |
Collapse
|
15
|
Sutanto H, Soegiarto G. Risk of Thrombosis during and after a SARS-CoV-2 Infection: Pathogenesis, Diagnostic Approach, and Management. Hematol Rep 2023; 15:225-243. [PMID: 37092518 PMCID: PMC10123679 DOI: 10.3390/hematolrep15020024] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/07/2023] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) increases the risk of thromboembolic events, especially in patients with severe infections requiring intensive care and cardiorespiratory support. COVID-19 patients with thromboembolic complications have a higher risk of death, and if they survive, these complications are expected to negatively affect these patients’ quality of life. Moreover, recent data reported that the risk of thromboembolism remains high months after a COVID-19 infection. Therefore, understanding the pathogenesis of thrombosis in the setting of COVID-19 may facilitate the early prevention and treatment of COVID-19-associated thromboembolism to reduce concomitant morbidity, mortality, and disability. This review will first discuss the clinical characteristics of COVID-19 infections, particularly with regard to the underlying pathophysiology. Then, the pathogenesis of COVID-19-associated thrombosis at the molecular and cellular levels will be comprehensively reviewed. Next, the clinical manifestations of venous and arterial thromboembolism in COVID-19 as well as the potential benefits of several laboratory markers of thrombosis will be further discussed. Lastly, the preventive and therapeutic management of thromboembolism during and after COVID-19 will also be explained.
Collapse
Affiliation(s)
- Henry Sutanto
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Department of Internal Medicine, Dr. Soetomo Teaching Hospital, Surabaya 60286, Indonesia
| | - Gatot Soegiarto
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Department of Internal Medicine, Dr. Soetomo Teaching Hospital, Surabaya 60286, Indonesia
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| |
Collapse
|
16
|
Hulme J. COVID-19 and Diarylamidines: The Parasitic Connection. Int J Mol Sci 2023; 24:6583. [PMID: 37047556 PMCID: PMC10094973 DOI: 10.3390/ijms24076583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
As emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants (Omicron) continue to outpace and negate combinatorial vaccines and monoclonal antibody therapies targeting the spike protein (S) receptor binding domain (RBD), the appetite for developing similar COVID-19 treatments has significantly diminished, with the attention of the scientific community switching to long COVID treatments. However, treatments that reduce the risk of "post-COVID-19 syndrome" and associated sequelae remain in their infancy, particularly as no established criteria for diagnosis currently exist. Thus, alternative therapies that reduce infection and prevent the broad range of symptoms associated with 'post-COVID-19 syndrome' require investigation. This review begins with an overview of the parasitic-diarylamidine connection, followed by the renin-angiotensin system (RAS) and associated angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSSR2) involved in SARS-CoV-2 infection. Subsequently, the ability of diarylamidines to inhibit S-protein binding and various membrane serine proteases associated with SARS-CoV-2 and parasitic infections are discussed. Finally, the roles of diarylamidines (primarily DIZE) in vaccine efficacy, epigenetics, and the potential amelioration of long COVID sequelae are highlighted.
Collapse
Affiliation(s)
- John Hulme
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si 461-701, Republic of Korea
| |
Collapse
|