408 results on '"Nafamostat"'
Search Results
2. Single and Multi-Dose Study, Pharmacokinetics of Oxycodone and PF614 Co-Administered with Nafamostat (PF614-MPAR-102)
- Author
-
Quotient Sciences
- Published
- 2024
3. Oral Nafamostat in Healthy Volunteers (NAF-101)
- Author
-
National Institute on Drug Abuse (NIDA)
- Published
- 2024
4. Pharmacokinetics of Oxycodone and PF614 Co-Administered with Nafamostat (PF614-MPAR-101) (MPAR-101)
- Author
-
Quotient Sciences and National Institute on Drug Abuse (NIDA)
- Published
- 2024
5. The Safety of Nafamostat Mesylate for Patients With High Risk Bleeding Undergoing Hemodialysis: A Pilot Study (Nafamostat)
- Published
- 2024
6. Efficacy and Safety Evaluation of Treatment Regimens in Adult COVID-19 Patients in Senegal (SEN-CoV-Fadj)
- Author
-
Fann Hospital, Senegal, Ministry of Health, Senegal, Diamniadio Children Hospital, Senegal, Dalal Jamm Hospital, Senegal, and Institut Pasteur Korea
- Published
- 2024
7. Australasian COVID-19 Trial (ASCOT) ADAptive Platform Trial (ASCOT)
- Author
-
The Peter Doherty Institute for Infection and Immunity, Australasian Society for Infectious Diseases, Hunter Medical Research Institute, Aotearoa Clinical Trials, Australian and New Zealand Intensive Care Research Centre, and Associate Professor Steven Tong, Associate Professor
- Published
- 2024
8. Comparison of Nafamostat and Unfractionated Heparin in RRT for Sepsis Associated AKI
- Author
-
Shen Lei, Chief Physician
- Published
- 2024
9. Pathophysiological dynamics in the contact, coagulation, and complement systems during sepsis: Potential targets for nafamostat mesilate
- Author
-
Qiaolan He, Yilin Wei, Yiqi Qian, and Ming Zhong
- Subjects
Sepsis ,Nafamostat ,Contact system ,Coagulation system ,Complement system ,Medical emergencies. Critical care. Intensive care. First aid ,RC86-88.9 - Abstract
Sepsis is a life-threatening syndrome resulting from a dysregulated host response to infection. It is the primary cause of death in the intensive care unit, posing a substantial challenge to human health and medical resource allocation. The pathogenesis and pathophysiology of sepsis are complex. During its onset, pro-inflammatory and anti-inflammatory mechanisms engage in intricate interactions, possibly leading to hyperinflammation, immunosuppression, and long-term immune disease. Of all critical outcomes, hyperinflammation is the main cause of early death among patients with sepsis. Therefore, early suppression of hyperinflammation may improve the prognosis of these patients. Nafamostat mesilate is a serine protease inhibitor, which can inhibit the activation of the complement system, coagulation system, and contact system. In this review, we discuss the pathophysiological changes occurring in these systems during sepsis, and describe the possible targets of the serine protease inhibitor nafamostat mesilate in the treatment of this condition.
- Published
- 2024
- Full Text
- View/download PDF
10. Use of Nafamostat Mesilate for Anticoagulation in Patients With ECMO
- Author
-
Xiaotong Hou, Clinical professor
- Published
- 2024
11. TMPRSS2 inhibitors for the treatment of COVID-19 in adults: a systematic review and meta-analysis of randomized clinical trials of nafamostat and camostat mesylate.
- Author
-
Hernández-Mitre, María Patricia, Morpeth, Susan C., Venkatesh, Balasubramanian, Hills, Thomas E., Davis, Joshua, Mahar, Robert K., McPhee, Grace, Jones, Mark, Totterdell, James, Tong, Steven Y.C., and Roberts, Jason A.
- Subjects
- *
COVID-19 treatment , *CLINICAL trials , *ADULTS , *SCIENCE databases , *MORTALITY - Abstract
Synthetic serine protease inhibitors block the cellular enzyme transmembrane protease serine 2, thus preventing SARS-CoV-2 cell entry. There are two relevant drugs in this class, namely, nafamostat (intravenous formulation) and camostat (oral formulation). To determine whether transmembrane protease serine 2 inhibition with nafamostat or camostat is associated with a reduced risk of 30-day all-cause mortality in adults with COVID-19. Scientific databases and clinical trial registry platforms. Preprints or published randomized clinical trials (RCTs) of nafamostat or camostat vs. usual care or placebo in adults requiring treatment for COVID-19. The primary outcome of the meta-analysis was 30-day all-cause mortality. Secondary outcomes included time to recovery, adverse events, and serious adverse events. Risk of bias (RoB) was assessed using the revised Cochrane RoB 2 tool for individually randomized trials. Meta-analysis was conducted in the R package meta (v7.0-0) using inverse variance and random effects. Protocol registration number was INPLASY202320120. Twelve RCTs were included. Overall, the number of available patients was small (nafamostat = 387; camostat = 1061), the number of enrolled patients meeting the primary outcome was low (nafamostat = 12; camostat = 13), and heterogeneity was high. In hospitalized adults, we did not identify differences in 30-day all-cause mortality (risk ratio [95% CI]: 0.58 [0.19, 1.80], p 0.34; I 2 = 0%; n = 6) and time to recovery (mean difference [95% CI]: 0.08 days [−0.74, 0.89], p 0.86; n = 2) between nafamostat vs. usual care; and for 30-day all-cause mortality (risk ratio [95% CI]: 0.99 [0.31, 3.18], p 0.99; n = 2) between camostat vs. placebo. The RCT evidence is inconclusive to determine whether there is a mortality reduction and safety with either nafamostat or camostat for the treatment of adults with COVID-19. There were high RoB, small sample size, and high heterogeneity between RCTs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. DEFINE - Evaluating Therapies for COVID-19 (DEFINE)
- Author
-
University of Oxford, Latus Therapeutics, and Scottish National Blood Transfusion Service (SNBTS)
- Published
- 2023
13. Efficacy of Nafamostat in Covid-19 Patients (RACONA Study) (RACONA)
- Author
-
Yokohama City University, University of Zurich, and Gian Paolo Rossi, MD, FAHA, FACC, Prof.
- Published
- 2023
14. Nafamostat Mesilate in the Treatment of Severe Infection-associated Coagulopathy (NMSICRCT)
- Author
-
Tianjin Third Central Hospital, Tianjin Medical University General Hospital, Shanxi Provincial People's Hospital, Shanxi Bethune Hospital, Hohhot First Hospital, The First Bethune Hospital Jilin University, Northern Jiangsu People's Hospital, The Affiliated Hospital of Nantong University, The Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, The First Affiliated Hospital of Suzhou Medical, Anhui Provincial Hospita, Zhejiang Hospital, Zhejiang Provincial People's Hospital, The First Affiliated Hospital of Ningbo, The First Affiliated Hospital of Wenzhou, The Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Gannan Medical University, The Second Affiliated Hospital Xi'an Jiaotong, LanZhou University, The First Affiliated Hospital of Xinjiang, First Affiliated Hospital of Xinjiang Medical University, Tongji Medical College of Huazhong University, The Second Hospital University of South China, The Fourth Affiliated Hospital of China Medical, The First Affiliated Hospital of Liaoning, Genertec Liaoyou Gem Flower Hospital, The Tenth People's Hospital of Shenyang, The Sixth People's Hospital of Shenyang, Dalian NO.3 People's Hospital, Benxi Cental Hospital, Liaoyang City Central Hospital, Huludao central hospital, and Xu Li, Professor
- Published
- 2023
15. Efficacy and Safety of Nafamostat Mesylate for VV-ECMO Anticoagulation
- Author
-
Xiaobo Yang, MD, Clinical Professor
- Published
- 2023
16. Drug repurposing for the treatment of COVID-19: Targeting nafamostat to the lungs by a liposomal delivery system.
- Author
-
Reus, Philipp, Guthmann, Hadar, Uhlig, Nadja, Agbaria, Majd, Issmail, Leila, Eberlein, Valentina, Nordling-David, Mirjam M., Jbara-Agbaria, Doaa, Ciesek, Sandra, Bojkova, Denisa, Cinatl, Jindrich, Burger-Kentischer, Anke, Rupp, Steffen, Zaliani, Andrea, Grunwald, Thomas, Gribbon, Philip, Kannt, Aimo, and Golomb, Gershon
- Subjects
- *
DRUG repositioning , *LUNGS , *COVID-19 treatment , *SARS-CoV-2 Omicron variant , *INHALATION administration , *INTRANASAL administration - Abstract
Despite tremendous global efforts since the beginning of the COVID-19 pandemic, still only a limited number of prophylactic and therapeutic options are available. Although vaccination is the most effective measure in preventing morbidity and mortality, there is a need for safe and effective post-infection treatment medication. In this study, we explored a pipeline of 21 potential candidates, examined in the Calu-3 cell line for their antiviral efficacy, for drug repurposing. Ralimetinib and nafamostat, clinically used drugs, have emerged as attractive candidates. Due to the inherent limitations of the selected drugs, we formulated targeted liposomes suitable for both systemic and intranasal administration. Non-targeted and targeted nafamostat liposomes (LipNaf) decorated with an Apolipoprotein B peptide (ApoB-P) as a specific lung-targeting ligand were successfully developed. The developed liposomal formulations of nafamostat were found to possess favorable physicochemical properties including nano size (119–147 nm), long-term stability of the normally rapidly degrading compound in aqueous solution, negligible leakage from the liposomes upon storage, and a neutral surface charge with low polydispersity index (PDI). Both nafamostat and ralimetinib liposomes showed good cellular uptake and lack of cytotoxicity, and non-targeted LipNaf demonstrated enhanced accumulation in the lungs following intranasal (IN) administration in non-infected mice. LipNaf retained its anti-SARS-CoV 2 activity in Calu 3 cells with only a modest decrease, exhibiting complete inhibition at concentrations >100 nM. IN, but not intraperitoneal (IP) treatment with targeted LipNaf resulted in a trend to reduced viral load in the lungs of K18-hACE2 mice compared to targeted empty Lip. Nevertheless, upon removal of outlier data, a statistically significant 1.9-fold reduction in viral load was achieved. This observation further highlights the importance of a targeted delivery into the respiratory tract. In summary, we were able to demonstrate a proof-of-concept of drug repurposing by liposomal formulations with anti-SARS-CoV-2 activity. The biodistribution and bioactivity studies with LipNaf suggest an IN or inhalation route of administration for optimal therapeutic efficacy. [Display omitted] • Drug repurposing for COVID-19 therapy: Liposomal delivery of ralimetinib/nafamostat. • Inhibition of different SARS-CoV-2 variants including omicron. • Liposomal delivery system suitable for both systemic and intranasal administration. • Apo-B peptide decorated liposomes: Targeted delivery to proteoglycans and LDLr. • Marked virus inhibition in SARS-CoV-2 infected mice, without side effects. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. Low-dose nafamostat mesilate ameliorates tissue injury and inhibits 5-hydroxytryptamine synthesis in the rat intestine after methotrexate administration
- Author
-
Takahiro Yamamoto, Takuji Machida, Chiho Tanno, Shiori Hasebe, Mayu Tamura, Nanaka Kobayashi, Sachiko Hiraide, Naoya Hamaue, and Kenji Iizuka
- Subjects
Methotrexate ,Nafamostat ,Gastrointestinal injury ,5-Hydroxytryptamine ,Enterochromaffin cells ,Therapeutics. Pharmacology ,RM1-950 - Abstract
We aimed to clarify the effect of nafamostat mesilate (nafamostat) on intestinal mucositis as well as the potentiation of intestinal 5-hydroxytryptamine (5-HT) dynamics induced by methotrexate, an anti-cancer drug, in rats. Rats received intraperitoneal methotrexate at 12.5 mg/kg/day for 4 days. In addition, 1, 3, or 10 mg/kg/day of nafamostat was given subcutaneously for 4 days. Ninety-six hours after the first administration of methotrexate, jejunal tissues were collected for analysis. The results showed that 1 mg/kg, but not 3 or 10 mg/kg, of nafamostat significantly ameliorated the methotrexate-induced body weight loss. Moreover, 1 mg/kg of nafamostat significantly improved methotrexate-induced mucositis, including villus atrophy. Nafamostat (1 mg/kg) significantly inhibited the methotrexate-induced mRNA expression of pro-inflammatory cytokines and cyclooxygenase-2, as well as methotrexate-induced 5-HT content and tryptophan hydroxylase (TPH) activity. In addition, it tended to inhibit the number of anti-TPH antibody-positive cells and significantly inhibited the number of anti-substance P antibody-positive cells. These findings suggest that low-dose nafamostat ameliorates tissue injury and 5-HT and substance P synthesis in methotrexate-induced mucositis. Nafamostat may be a novel therapeutic strategy for the prevention and treatment of mucositis as well as 5-HT- and/or substance P-related adverse effects in cancer chemotherapy.
- Published
- 2023
- Full Text
- View/download PDF
18. Mechanisms of NMDA Receptor Inhibition by Sepimostat—Comparison with Nafamostat and Diarylamidine Compounds.
- Author
-
Zhigulin, Arseniy S. and Barygin, Oleg I.
- Subjects
- *
GUANIDINES , *PYRAMIDAL neurons , *BINDING constant , *BINDING sites , *PROTEASE inhibitors , *METHYL aspartate receptors , *METHYL aspartate - Abstract
N-methyl-D-aspartate (NMDA) receptors are inhibited by many amidine and guanidine compounds. In this work, we studied the mechanisms of their inhibition by sepimostat—an amidine-containing serine protease inhibitor with neuroprotective properties. Sepimostat inhibited native NMDA receptors in rat hippocampal CA1 pyramidal neurons with IC50 of 3.5 ± 0.3 µM at −80 mV holding voltage. It demonstrated complex voltage dependence with voltage-independent and voltage-dependent components, suggesting the presence of shallow and deep binding sites. At −80 mV holding voltage, the voltage-dependent component dominates, and we observed pronounced tail currents and overshoots evidencing a "foot-in-the-door" open channel block. At depolarized voltages, the voltage-independent inhibition by sepimostat was significantly attenuated by the increase of agonist concentration. However, the voltage-independent inhibition was non-competitive. We further compared the mechanisms of the action of sepimostat with those of structurally-related amidine and guanidine compounds—nafamostat, gabexate, furamidine, pentamidine, diminazene, and DAPI—investigated previously. The action of all these compounds can be described by the two-component mechanism. All compounds demonstrated similar affinity to the shallow site, which is responsible for the voltage-independent inhibition, with binding constants in the range of 3–30 µM. In contrast, affinities to the deep site differed dramatically, with nafamostat, furamidine, and pentamidine being much more active. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
19. Conformational response to ligand binding of TMPRSS2, a protease involved in SARS‐CoV‐2 infection: Insights through computational modeling.
- Author
-
Frumenzio, Giorgia, Chandramouli, Balasubramanian, Besker, Neva, Grottesi, Alessandro, Talarico, Carmine, Frigerio, Francesco, Emerson, Andrew, and Musiani, Francesco
- Abstract
Thanks to the considerable research which has been undertaken in the last few years to improve our understanding of the biology and mechanism of action of SARS‐CoV‐2, we know how the virus uses its surface spike protein to infect host cells. The transmembrane prosthesis, serine 2 (TMPRSS2) protein, located on the surface of human cells, recognizes the cleavage site in the spike protein, leading to the release of the fusion peptide and entry of the virus into the host cells. Because of its role, TMPRSS2 has been proposed as a drug target to prevent infection by the virus. In this study, we aim to increase our understanding of TMPRSS2 using long scale microsecond atomistic molecular dynamics simulations, focusing on the conformational changes over time. The comparison between simulations conducted on the protein in the native (apo) and inhibited form (holo), has shown that in the holo form the inhibitor stabilizes the catalytic site and induces rearrangements in the extracellular domain of the protein. In turn, it leads to the formation of a new cavity in the vicinity of the ligand binding pocket that is stable in the microsecond time scale. Given the low specificity of known protease inhibitors, these findings suggest a new potential drug target site that can be used to improve TMPRSS2 specific recognition by newly designed inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
20. An autopsy case of a patient on maintenance hemodialysis with continuous idiopathic cholesterol crystal embolism for 7 years
- Author
-
Yumiko Kawai, Chiharu Kinoshita, Koichi Matsuda, Keiko Takeuchi, Yoshimoto Inoue, and Yoko Fujita
- Subjects
Cholesterol crystal embolism ,Idiopathic cholesterol crystal embolism ,Autopsy ,Maintenance hemodialysis ,Nafamostat ,Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
Abstract Background CCE is a systemic disease with poor prognosis with no established treatment. Approximately 23–32% of CCE cases progress to end-stage renal failure, and the 1-year mortality rate of CCE with organ failure is 60–90%. The dialysis method for the patients with CCE is still controversial. Case report The patient is 73 years old male who was diagnosed with idiopathic CCE. He had survived 7 years though he had been on maintenance HD. We used nafamostat for HD every time. He took prednisolone and statin. He died due to rupture of AAA and we autopsied him. CCs developed in five organs, including the right lung CCE was assumed to be continuously present since the diagnosis. Discussion and conclusion CCE was continuous until death, and CCs in the right lung were possibly due to HD. HD through AV shunt could worsen CCE, and HD should be recognized as the aggravating factor. The use of nafamostat while undergoing HD as well as use of steroids and statins until death may have prevented fatal events and contributed to the patient's long survival.
- Published
- 2023
- Full Text
- View/download PDF
21. Evaluation of Nafamostat as Chemoprophylaxis for SARS-CoV-2 Infection in Hamsters.
- Author
-
Neary, Megan, Sharp, Joanne, Gallardo-Toledo, Eduardo, Herriott, Joanne, Kijak, Edyta, Bramwell, Chloe, Cox, Helen, Tatham, Lee, Box, Helen, Curley, Paul, Arshad, Usman, Rajoli, Rajith K. R., Pertinez, Henry, Valentijn, Anthony, Dhaliwal, Kevin, Mc Caughan, Frank, Hobson, James, Rannard, Steve, Kipar, Anja, and Stewart, James P.
- Subjects
- *
SARS-CoV-2 , *CHEMOPREVENTION , *HAMSTERS , *LUNGS , *TURBINATE bones , *GOLDEN hamster , *NASAL mucosa - Abstract
The successful development of a chemoprophylaxis against SARS-CoV-2 could provide a tool for infection prevention that is implementable alongside vaccination programmes. Nafamostat is a serine protease inhibitor that inhibits SARS-CoV-2 entry in vitro, but it has not been characterised for chemoprophylaxis in animal models. Clinically, nafamostat is limited to intravenous delivery and has an extremely short plasma half-life. This study sought to determine whether intranasal dosing of nafamostat at 5 mg/kg twice daily was able to prevent the airborne transmission of SARS-CoV-2 from infected to uninfected Syrian Golden hamsters. SARS-CoV-2 RNA was detectable in the throat swabs of the water-treated control group 4 days after cohabitation with a SARS-CoV-2 inoculated hamster. However, throat swabs from the intranasal nafamostat-treated hamsters remained SARS-CoV-2 RNA negative for the full 4 days of cohabitation. Significantly lower SARS-CoV-2 RNA concentrations were seen in the nasal turbinates of the nafamostat-treated group compared to the control (p = 0.001). A plaque assay quantified a significantly lower concentration of infectious SARS-CoV-2 in the lungs of the nafamostat-treated group compared to the control (p = 0.035). When taken collectively with the pathological changes observed in the lungs and nasal mucosa, these data are strongly supportive of the utility of intranasally delivered nafamostat for the prevention of SARS-CoV-2 infection. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
22. Evaluation of the Antiviral Efficacy of Subcutaneous Nafamostat Formulated with Glycyrrhizic Acid against SARS-CoV-2 in a Murine Model.
- Author
-
Jeong, Ju Hwan, Lee, Woong Hee, Min, Seong Cheol, Kim, Beom Kyu, Park, On Bi, Chokkakula, Santosh, Ahn, Seong Ju, Oh, Sol, Park, Ji-Hyun, Jung, Ji Won, Jung, Ji Min, Kim, Eung-Gook, and Song, Min-Suk
- Subjects
- *
SARS-CoV-2 , *ANTIVIRAL agents , *DRUG repositioning , *SPRAGUE Dawley rats , *TRANSGENIC mice , *LABORATORY mice - Abstract
The ongoing COVID-19 pandemic highlights the urgent need for effective antiviral agents and vaccines. Drug repositioning, which involves modifying existing drugs, offers a promising approach for expediting the development of novel therapeutics. In this study, we developed a new drug, MDB-MDB-601a-NM, by modifying the existing drug nafamostat (NM) with the incorporation of glycyrrhizic acid (GA). We assessed the pharmacokinetic profiles of MDB-601a-NM and nafamostat in Sprague-Dawley rats, revealing rapid clearance of nafamostat and sustained drug concentration of MDB-601a-NM after subcutaneous administration. Single-dose toxicity studies showed potential toxicity and persistent swelling at the injection site with high-dose administration of MDB-601a-NM. Furthermore, we evaluated the efficacy of MDB-601a-NM in protecting against SARS-CoV-2 infection using the K18 hACE-2 transgenic mouse model. Mice treated with 60 mg/kg and 100 mg/kg of MDB-601a-NM exhibited improved protectivity in terms of weight loss and survival rates compared to the nafamostat-treated group. Histopathological analysis revealed dose-dependent improvements in histopathological changes and enhanced inhibitory efficacy in MDB-601a-NM-treated groups. Notably, no viral replication was detected in the brain tissue when mice were treated with 60 mg/kg and 100 mg/kg of MDB-601a-NM. Our developed MDB-601a-NM, a modified Nafamostat with glycyrrhizic acid, shows improved protectivity against SARS-CoV-2 infection. Its sustained drug concentration after subcutaneous administration and dose-dependent improvements makes it a promising therapeutic option. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
23. Characteristics of pediatric continuous renal replacement therapies in hospitals with pediatric intensive care units in Japan.
- Author
-
Haga, Taiki, Ide, Kentaro, and Tani, Masanori
- Subjects
RENAL replacement therapy ,PEDIATRIC intensive care ,CHILDREN'S hospitals ,INTENSIVE care units ,PEDIATRIC therapy - Abstract
Introduction: Information on the implementation of continuous renal replacement therapy (CRRT) in pediatric intensive care units (PICUs) is limited. We investigated the real‐world practice of this therapy in Japan. Methods: We conducted a cross‐sectional survey of 26 hospitals with PICUs in Japan. One physician per hospital responded to the questionnaire. Results: Fewer than half of the hospitals followed the CRRT practice guidelines; treatment options were often selected at a physician's discretion. PICUs varied widely in continuous renal replacement settings, circuit priming methods, frequency of circuit exchange, and policies for using CRRT in sepsis. Only two anticoagulants were used: nafamostat mesylate and unfractionated heparin. Most units did not change the nutritional dosage according to CRRT use. Many facilities were proactive concerning rehabilitation. Conclusion: Nafamostat‐centered anticoagulation management was unique to Japan. Our results may help identify areas where additional clinical studies are needed to standardize CRRT practice. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
24. A Study Evaluating the Efficacy and Safety of CKD-314 in Hospitalized Adult Patients Diagnosed With COVID-19 Pneumonia
- Published
- 2021
25. Phase 3 Clinical Trial to Evaluate the Efficacy and Safety of CKD-314
- Published
- 2021
26. A Study Evaluating the Efficacy and Safety of CKD-314 (Nafabelltan) in Hospitalized Adult Patients Diagnosed With COVID-19 Pneumonia
- Published
- 2021
27. TCT‐Mediated Process for the Preparation of Nafamostat Mesylate and Camostat Mesylate.
- Author
-
Ahmed, Riyaz, Kumar, Gulshan, Mahajan, Sheena, Verma, Praveen Kumar, Singh Cham, Pankaj, Reddy, D. Srinivasa, Shankar, Ravi, and Singh, Parvinder Pal
- Subjects
- *
CYANURIC acid , *ACID throwing , *HYDROXYL group , *RAW materials , *PYRIDINE - Abstract
The present work describes the s‐trichlorotriazine (TCT)‐mediated formation of highly functionalized ester‐based drugs, namely nafamostat mesylate and camostat mesylate. The TCT‐mediated activation of acids followed by the attack of a hydroxyl group (−OH group) resulted in the formation of ester. The high aqueous solubility of cyanuric acid eases the work‐up process and avoids repetitive washing process (better E‐factor). The present method also avoids using pyridine as a base, and instead of that, N‐methylmorpholine (NMM) was used for the coupling. Moreover, we have also demonstrated the synthesis of key intermediates namely, 4‐guanidinobenzoic acid and 6‐hydroxy‐2‐naphthimidamide from economical and readily available raw materials. The TCT‐mediated coupling provides many advantages in scalability, cost‐effectiveness and environmental friendly handling. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
28. Nafamostat has anti-asthmatic effects associated with suppressed pro-inflammatory gene expression, eosinophil infiltration and airway hyperreactivity.
- Author
-
Raju Allam, Venkata Sita Rama, Waern, Ida, Taha, Sowsan, Akula, Srinivas, Wernersson, Sara, and Pejler, Gunnar
- Subjects
GENE expression ,HOUSE dust mites ,EOSINOPHILS ,AIRWAY (Anatomy) ,MAST cells - Abstract
Introduction: Asthma is characterized by an imbalance between proteases and their inhibitors. Hence, an attractive therapeutic option could be to interfere with asthma-associated proteases. Here we exploited this option by assessing the impact of nafamostat, a serine protease inhibitor known to neutralize mast cell tryptase. Methods: Nafamostat was administered in a mouse model for asthma based on sensitization by house dust mite (HDM) extract, followed by the assessment of effects on airway hyperreactivity, inflammatory parameters and gene expression. Results: We show that nafamostat efficiently suppressed the airway hyperreactivity in HDM-sensitized mice. This was accompanied by reduced infiltration of eosinophils and lymphocytes to the airways, and by lower levels of pro-inflammatory compounds within the airway lumen. Further, nafamostat had a dampening impact on goblet cell hyperplasia and smooth muscle layer thickening in the lungs of HDM-sensitized animals. To obtain deeper insight into the underlying mechanisms, a transcriptomic analysis was conducted. This revealed, as expected, that the HDM sensitization caused an upregulated expression of numerous pro-inflammatory genes. Further, the transcriptomic analysis showed that nafamostat suppressed the levels of multiple proinflammatory genes, with a particular impact on genes related to asthma. Discussion: Taken together, this study provides extensive insight into the ameliorating effect of nafamostat on experimental asthma, and our findings can thereby provide a basis for the further evaluation of nafamostat as a potential therapeutic agent in human asthma. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
29. Inhibitory potentials of ivermectin, nafamostat, and camostat on spike protein and some nonstructural proteins of SARS-CoV-2: Virtual screening approach
- Author
-
Haruna Isiyaku Umar, Ijeoma Akunna Duru, Uchechi Emmanuela Enenebeaku, Lynda Chioma Ngozi Olehi, Christian Ebere Enyoh, and Chidi Edbert Duru
- Subjects
camostat ,ivermectin ,nafamostat ,nonstructural protein ,spike protein ,virtual screening ,Medicine (General) ,R5-920 - Abstract
The search for potent oral drugs either through synthetic routes or by drug repurposing for combating the dreaded covid-19 virus is still ongoing. The coronavirus spike glycoprotein and several other non-structural proteins play crucial roles in the replication and transmission of this virus. Recent research have identified ivermectin, nafamostat, and camostat as promising drug inhibitors of SARS-CoV-2 target proteins. The broad-spectrum inhibitory action of ivermectin, nafamostat, and camostat on the spike glycoprotein and some non-structural proteins of this virus was studied in silico. The spike glycoprotein, nsp3, nsp5, nsp9, nsp10, nsp13, and nsp16 were selected for this study and were downloaded from the protein data bank. Flexible docking procedure implemented in Auto Dock Vina module was deployed for the docking procedure of the drugs with the protein receptors. Although ivermectin had the best inhibitory action on the viral spike protein and nsp10, nafamostat was identified as the compound with the best broad-spectrum activity on this virus, having the highest binding affinity values of – 9.4kcal/mol, – 7.9 Kcal/mol, – 6.1 Kcal/mol, – 8.0 Kcal/mol, and – 8.7 Kcal/mol for nsp3, nsp5, nsp9, nsp13, and nsp16 respectively. This drug, in combination with ivermectin could therefore be explored further as potential compounds that could be modified to curb the menace of the covid-19 pandemic.
- Published
- 2022
- Full Text
- View/download PDF
30. Clinical Efficacy of Nafamostat Mesylate for COVID-19 Pneumonia
- Author
-
IN-GYU BAE, MD, Professor
- Published
- 2020
31. Nafamostat has anti-asthmatic effects associated with suppressed pro-inflammatory gene expression, eosinophil infiltration and airway hyperreactivity
- Author
-
Venkata Sita Rama Raju Allam, Ida Waern, Sowsan Taha, Srinivas Akula, Sara Wernersson, and Gunnar Pejler
- Subjects
nafamostat ,serine proteases ,asthma ,house dust mite ,cytokines ,airway hyperreactivity ,Immunologic diseases. Allergy ,RC581-607 - Abstract
IntroductionAsthma is characterized by an imbalance between proteases and their inhibitors. Hence, an attractive therapeutic option could be to interfere with asthma-associated proteases. Here we exploited this option by assessing the impact of nafamostat, a serine protease inhibitor known to neutralize mast cell tryptase.MethodsNafamostat was administered in a mouse model for asthma based on sensitization by house dust mite (HDM) extract, followed by the assessment of effects on airway hyperreactivity, inflammatory parameters and gene expression.ResultsWe show that nafamostat efficiently suppressed the airway hyperreactivity in HDM-sensitized mice. This was accompanied by reduced infiltration of eosinophils and lymphocytes to the airways, and by lower levels of pro-inflammatory compounds within the airway lumen. Further, nafamostat had a dampening impact on goblet cell hyperplasia and smooth muscle layer thickening in the lungs of HDM-sensitized animals. To obtain deeper insight into the underlying mechanisms, a transcriptomic analysis was conducted. This revealed, as expected, that the HDM sensitization caused an upregulated expression of numerous pro-inflammatory genes. Further, the transcriptomic analysis showed that nafamostat suppressed the levels of multiple pro-inflammatory genes, with a particular impact on genes related to asthma.DiscussionTaken together, this study provides extensive insight into the ameliorating effect of nafamostat on experimental asthma, and our findings can thereby provide a basis for the further evaluation of nafamostat as a potential therapeutic agent in human asthma.
- Published
- 2023
- Full Text
- View/download PDF
32. Predicting the systemic exposure and lung concentration of nafamostat using physiologically-based pharmacokinetic modeling.
- Author
-
Hyeon-Cheol Jeong, Yoon-Jee Chae, and Kwang-Hee Shin
- Subjects
- *
SARS-CoV-2 , *LUNGS , *COVID-19 , *PHARMACOKINETICS - Abstract
Nafamostat has been actively studied for its neuroprotective activity and effect on various indications, such as coronavirus disease 2019 (COVID-19). Nafamostat has low water solubility at a specific pH and is rapidly metabolized in the blood. Therefore, it is administered only intravenously, and its distribution is not well known. The main purposes of this study are to predict and evaluate the pharmacokinetic (PK) profiles of nafamostat in a virtual healthy population under various dosing regimens. The most important parameters were assessed using a physiologically based pharmacokinetic (PBPK) approach and global sensitivity analysis with the Sobol sensitivity analysis. A PBPK model was constructed using the SimCYP® simulator. Data regarding the in vitro metabolism and clinical studies were extracted from the literature to assess the predicted results. The model was verified using the arithmetic mean maximum concentration (Cmax), the area under the curve from 0 to the last time point (AUC0-t), and AUC from 0 to infinity (AUC0-8) ratio (predicted/observed), which were included in the 2-fold range. The simulation results suggested that the 2 dosing regimens for the treatment of COVID-19 used in the case reports could maintain the proposed effective concentration for inhibiting severe acute respiratory syndrome coronavirus 2 entry into the plasma and lung tissue. Global sensitivity analysis indicated that hematocrit, plasma half-life, and microsomal protein levels significantly influenced the systematic exposure prediction of nafamostat. Therefore, the PBPK modeling approach is valuable in predicting the PK profile and designing an appropriate dosage regimen. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
33. The effect of nafamostat mesilate infusion after ERCP for post-ERCP pancreatitis
- Author
-
Joo Seong Kim, Sang Hyub Lee, Namyoung Park, Gunn Huh, Jung Won Chun, Jin Ho Choi, In Rae Cho, Woo Hyun Paik, Ji Kon Ryu, and Yong-Tae Kim
- Subjects
Nafamostat ,Endoscopic retrograde cholangiopancreatography ,Post-ERCP pancreatitis ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Abstract Background Nafamostat mesilate decreases the incidence of pancreatitis after endoscopic retrograde cholangiopancreatography (ERCP). However, no studies have administered nafamostat mesilate after ERCP. So we investigated if the infusion of nafamostat mesilate after ERCP can affect the post-ERCP pancreatitis (PEP) in high-risk patients. Methods In a tertiary hospital, 350 high-risk patients of PEP were reviewed retrospectively. Among them, 201 patients received nafamostat mesilate after ERCP. Patient-related and procedure-related risk factors for PEP were collected. We performed a propensity score matching to adjust for the significant different baseline characteristics. The incidence and severity of PEP were evaluated according to the infusion of nafamostat mesilate. The risk factors of PEP were also analyzed by multivariate logistic regression. Results The baseline characteristics were not different after the matching. The PEP rate (17.4% vs. 10.3%, P = 0.141) was insignificant. Among the PEP patients, mild PEP was significantly higher in the nafamostat mesilate group (85.7% vs. 45.5%, P = 0.021). Only one patient in the nafamostat mesilate group developed severe PEP. Although young age (odds ratio [OR] 3.60, 95% CI 1.09–11.85, P = 0.035) was a risk factor, nafamostat mesilate (odds ratio [OR] 0.30, 95% CI 0.09–0.98, P = 0.047) was a protective factor for moderate to severe PEP. Conclusions The administration of nafamostat mesilate after ERCP in high-risk patients was not effective in preventing PEP, but may attenuate the severity of PEP.
- Published
- 2022
- Full Text
- View/download PDF
34. Mechanisms of NMDA Receptor Inhibition by Sepimostat—Comparison with Nafamostat and Diarylamidine Compounds
- Author
-
Arseniy S. Zhigulin and Oleg I. Barygin
- Subjects
NMDA receptors ,pharmacological modulation ,patch clamp ,sepimostat ,nafamostat ,gabexate ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
N-methyl-D-aspartate (NMDA) receptors are inhibited by many amidine and guanidine compounds. In this work, we studied the mechanisms of their inhibition by sepimostat—an amidine-containing serine protease inhibitor with neuroprotective properties. Sepimostat inhibited native NMDA receptors in rat hippocampal CA1 pyramidal neurons with IC50 of 3.5 ± 0.3 µM at −80 mV holding voltage. It demonstrated complex voltage dependence with voltage-independent and voltage-dependent components, suggesting the presence of shallow and deep binding sites. At −80 mV holding voltage, the voltage-dependent component dominates, and we observed pronounced tail currents and overshoots evidencing a “foot-in-the-door” open channel block. At depolarized voltages, the voltage-independent inhibition by sepimostat was significantly attenuated by the increase of agonist concentration. However, the voltage-independent inhibition was non-competitive. We further compared the mechanisms of the action of sepimostat with those of structurally-related amidine and guanidine compounds—nafamostat, gabexate, furamidine, pentamidine, diminazene, and DAPI—investigated previously. The action of all these compounds can be described by the two-component mechanism. All compounds demonstrated similar affinity to the shallow site, which is responsible for the voltage-independent inhibition, with binding constants in the range of 3–30 µM. In contrast, affinities to the deep site differed dramatically, with nafamostat, furamidine, and pentamidine being much more active.
- Published
- 2023
- Full Text
- View/download PDF
35. Broad antiviral and anti‐inflammatory efficacy of nafamostat against SARS‐CoV‐2 and seasonal coronaviruses in primary human bronchiolar epithelia
- Author
-
Brian F. Niemeyer, Caitlin M. Miller, Carmen Ledesma‐Feliciano, James H. Morrison, Rocio Jimenez‐Valdes, Clarissa Clifton, Eric M. Poeschla, and Kambez H. Benam
- Subjects
SARS‐CoV‐2 ,Nafamostat ,antiviral ,anti‐inflammatory ,airway epithelium ,Materials of engineering and construction. Mechanics of materials ,TA401-492 - Abstract
Abstract Antiviral strategies that target host systems needed for SARS‐CoV‐2 replication and pathogenesis may have therapeutic potential and help mitigate resistance development. Here, we evaluate nafamostat mesylate, a potent broad‐spectrum serine protease inhibitor that blocks host protease activation of the viral spike protein. SARS‐CoV‐2 is used to infect human polarized mucociliated primary bronchiolar epithelia reconstituted with cells derived from healthy donors, smokers and subjects with chronic obstructive pulmonary disease. Nafamostat markedly inhibits apical shedding of SARS‐CoV‐2 from all donors (log10 reduction). We also observe, for the first‐time, anti‐inflammatory effects of nafamostat on airway epithelia independent of its antiviral effects, suggesting a dual therapeutic advantage in the treatment of COVID‐19. Nafamostat also exhibits antiviral properties against the seasonal human coronaviruses 229E and NL6. These findings suggest therapeutic promise for nafamostat in treating SARS‐CoV‐2 and other human coronaviruses.
- Published
- 2022
- Full Text
- View/download PDF
36. Nafamostat protects against early brain injury after subarachnoid hemorrhage in mice
- Author
-
Hirofumi Matsubara, Takahiko Imai, Shohei Tsuji, Natsumi Oka, Yusuke Egashira, Yukiko Enomoto, Noriyuki Nakayama, Shinsuke Nakamura, Masamitsu Shimazawa, Toru Iwama, and Hideaki Hara
- Subjects
Nafamostat ,Subarachnoid hemorrhage ,Early brain injury ,Thrombin ,Therapeutics. Pharmacology ,RM1-950 - Abstract
This study aimed to evaluate the effects of nafamostat, a serin protease inhibitor, in the management of subarachnoid hemorrhage (SAH). SAH was induced by endovascular perforation in male mice. Nafamostat was administered intraperitoneally four times immediately after SAH induction. Cerebral blood flow, neurological behavior tests, SAH grade and protein expression were evaluated at 24 h after SAH induction. In the in vitro model, human brain microvascular endothelial cells (HBMVECs), HBVECs were exposed to thrombin and hypoxia for 24 h; nafamostat was administered and the protein expression was evaluated.Eighty-eight mice were included in the in vivo study. Fifteen mice (17%) were excluded because of death or procedure failure. Nafamostat exerted no significant effect on the SAH grade or cerebral blood flow; however, it improved the neurological behavior and suppressed the thrombin and MMP-9 expression. In addition, nafamostat suppressed the ICAM-1 expression and p38 phosphorylation in the in vitro study.Nafamostat has a protective effect against HBMVEC after exposure to thrombin and hypoxia, suggesting its role in improving the neurological outcomes after SAH. These findings indicate that nafamostat has the potential to be a novel therapeutic drug in the management of SAH.
- Published
- 2022
- Full Text
- View/download PDF
37. Nafamostat reduces systemic inflammation in TLR7-mediated virus-like illness
- Author
-
Abi G. Yates, Caroline M. Weglinski, Yuxin Ying, Isobel K. Dunstan, Tatyana Strekalova, and Daniel C. Anthony
- Subjects
Nafamostat ,Viral infection ,Inflammation ,Sickness behaviour ,COVID-19 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background The serine protease inhibitor nafamostat has been proposed as a treatment for COVID-19, by inhibiting TMPRSS2-mediated viral cell entry. Nafamostat has been shown to have other, immunomodulatory effects, which may be beneficial for treatment, however animal models of ssRNA virus infection are lacking. In this study, we examined the potential of the dual TLR7/8 agonist R848 to mimic the host response to an ssRNA virus infection and the associated behavioural response. In addition, we evaluated the anti-inflammatory effects of nafamostat in this model. Methods CD-1 mice received an intraperitoneal injection of R848 (200 μg, prepared in DMSO, diluted 1:10 in saline) or diluted DMSO alone, and an intravenous injection of either nafamostat (100 μL, 3 mg/kg in 5% dextrose) or 5% dextrose alone. Sickness behaviour was determined by temperature, food intake, sucrose preference test, open field and forced swim test. Blood and fresh liver, lung and brain were collected 6 h post-challenge to measure markers of peripheral and central inflammation by blood analysis, immunohistochemistry and qPCR. Results R848 induced a robust inflammatory response, as evidenced by increased expression of TNF, IFN-γ, CXCL1 and CXCL10 in the liver, lung and brain, as well as a sickness behaviour phenotype. Exogenous administration of nafamostat suppressed the hepatic inflammatory response, significantly reducing TNF and IFN-γ expression, but had no effect on lung or brain cytokine production. R848 administration depleted circulating leukocytes, which was restored by nafamostat treatment. Conclusions Our data indicate that R848 administration provides a useful model of ssRNA virus infection, which induces inflammation in the periphery and CNS, and virus infection-like illness. In turn, we show that nafamostat has a systemic anti-inflammatory effect in the presence of the TLR7/8 agonist. Therefore, the results indicate that nafamostat has anti-inflammatory actions, beyond its ability to inhibit TMPRSS2, that might potentiate its anti-viral actions in pathologies such as COVID-19.
- Published
- 2022
- Full Text
- View/download PDF
38. Anticoagulation with nafamostat mesilate during extracorporeal life support.
- Author
-
Lang, Yuheng, Zheng, Yue, Qi, Bingcai, Zheng, Weifeng, Wei, Jin, Zhao, Chengxiu, Gao, Wenqing, and Li, Tong
- Subjects
- *
HEART assist devices , *EXTRACORPOREAL membrane oxygenation , *CARDIOGENIC shock , *RENAL replacement therapy , *ANTICOAGULANTS , *T cells , *RESPIRATORY diseases - Abstract
Nafamostat mesylate (NM) affects coagulation and fibrinolysis and impedes obesity-associated protein demethylase activity, which regulates Na+/K+ transport properties and the NF-κB signaling pathway. NM significantly decreases macrophage, neutrophil, and T lymphocyte infiltration, thereby reducing inflammation and apoptosis after reperfusion and promoting recovery in patients with severe conditions such as near-fatal asthma and cardiac arrest. Extracorporeal life support (ECLS) devices are used for cardiac and/or pulmonary support as a bridge to recovery, decision, surgery, or transplant in patients with refractory cardio-circulatory or respiratory diseases and provide essential opportunities for organ support and patient survival. However, they can lead to some potential adverse events such as hemorrhage and thrombosis. NM provides a sustained innate immune response of coagulation and anti-inflammation in extracorporeal circuits, principally due to its activation of the contact and complement systems. Heparin is the main anticoagulant used in extracorporeal circuits; however, it may cause massive bleeding and heparin-induced thrombocytopenia. Although no antidote is available, NM has a very short half-life of approximately 8–10 min and might have positive effects on patients who require coagulation and anti-inflammation. NM has been used for anticoagulation in continuous renal replacement therapy, extracorporeal membrane oxygenation, hemodialysis, and left ventricular assist devices. In this review, we focused on the pharmacology, monitoring parameters, and considerations for the special use of NM in patients receiving ECLS. Our findings suggest that systemic anticoagulation with NM during ECLS might be a feasible and safe alternative with several advantages for critically ill patients with high-risk bleeding and might improve their prognosis. • Nafamostat Mesilate affects anticoagulation and reduce the risk of bleeding • Nafamostat Mesilate shown anti-inflammatory effects for patients with ECMO • Nafamostat Mesilate has a very short half-time compared to other anticoagulation drugs [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
39. An autopsy case of a patient on maintenance hemodialysis with continuous idiopathic cholesterol crystal embolism for 7 years
- Author
-
Kawai, Yumiko, Kinoshita, Chiharu, Matsuda, Koichi, Takeuchi, Keiko, Inoue, Yoshimoto, and Fujita, Yoko
- Published
- 2023
- Full Text
- View/download PDF
40. Evaluation of Nafamostat as Chemoprophylaxis for SARS-CoV-2 Infection in Hamsters
- Author
-
Megan Neary, Joanne Sharp, Eduardo Gallardo-Toledo, Joanne Herriott, Edyta Kijak, Chloe Bramwell, Helen Cox, Lee Tatham, Helen Box, Paul Curley, Usman Arshad, Rajith K. R. Rajoli, Henry Pertinez, Anthony Valentijn, Kevin Dhaliwal, Frank Mc Caughan, James Hobson, Steve Rannard, Anja Kipar, James P. Stewart, and Andrew Owen
- Subjects
SARS-CoV-2 ,nafamostat ,chemoprophylaxis ,Microbiology ,QR1-502 - Abstract
The successful development of a chemoprophylaxis against SARS-CoV-2 could provide a tool for infection prevention that is implementable alongside vaccination programmes. Nafamostat is a serine protease inhibitor that inhibits SARS-CoV-2 entry in vitro, but it has not been characterised for chemoprophylaxis in animal models. Clinically, nafamostat is limited to intravenous delivery and has an extremely short plasma half-life. This study sought to determine whether intranasal dosing of nafamostat at 5 mg/kg twice daily was able to prevent the airborne transmission of SARS-CoV-2 from infected to uninfected Syrian Golden hamsters. SARS-CoV-2 RNA was detectable in the throat swabs of the water-treated control group 4 days after cohabitation with a SARS-CoV-2 inoculated hamster. However, throat swabs from the intranasal nafamostat-treated hamsters remained SARS-CoV-2 RNA negative for the full 4 days of cohabitation. Significantly lower SARS-CoV-2 RNA concentrations were seen in the nasal turbinates of the nafamostat-treated group compared to the control (p = 0.001). A plaque assay quantified a significantly lower concentration of infectious SARS-CoV-2 in the lungs of the nafamostat-treated group compared to the control (p = 0.035). When taken collectively with the pathological changes observed in the lungs and nasal mucosa, these data are strongly supportive of the utility of intranasally delivered nafamostat for the prevention of SARS-CoV-2 infection.
- Published
- 2023
- Full Text
- View/download PDF
41. Evaluation of the Antiviral Efficacy of Subcutaneous Nafamostat Formulated with Glycyrrhizic Acid against SARS-CoV-2 in a Murine Model
- Author
-
Ju Hwan Jeong, Woong Hee Lee, Seong Cheol Min, Beom Kyu Kim, On Bi Park, Santosh Chokkakula, Seong Ju Ahn, Sol Oh, Ji-Hyun Park, Ji Won Jung, Ji Min Jung, Eung-Gook Kim, and Min-Suk Song
- Subjects
SARS-CoV-2 ,antiviral ,nafamostat ,glycyrrhizic acid ,in vivo mouse model ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
The ongoing COVID-19 pandemic highlights the urgent need for effective antiviral agents and vaccines. Drug repositioning, which involves modifying existing drugs, offers a promising approach for expediting the development of novel therapeutics. In this study, we developed a new drug, MDB-MDB-601a-NM, by modifying the existing drug nafamostat (NM) with the incorporation of glycyrrhizic acid (GA). We assessed the pharmacokinetic profiles of MDB-601a-NM and nafamostat in Sprague-Dawley rats, revealing rapid clearance of nafamostat and sustained drug concentration of MDB-601a-NM after subcutaneous administration. Single-dose toxicity studies showed potential toxicity and persistent swelling at the injection site with high-dose administration of MDB-601a-NM. Furthermore, we evaluated the efficacy of MDB-601a-NM in protecting against SARS-CoV-2 infection using the K18 hACE-2 transgenic mouse model. Mice treated with 60 mg/kg and 100 mg/kg of MDB-601a-NM exhibited improved protectivity in terms of weight loss and survival rates compared to the nafamostat-treated group. Histopathological analysis revealed dose-dependent improvements in histopathological changes and enhanced inhibitory efficacy in MDB-601a-NM-treated groups. Notably, no viral replication was detected in the brain tissue when mice were treated with 60 mg/kg and 100 mg/kg of MDB-601a-NM. Our developed MDB-601a-NM, a modified Nafamostat with glycyrrhizic acid, shows improved protectivity against SARS-CoV-2 infection. Its sustained drug concentration after subcutaneous administration and dose-dependent improvements makes it a promising therapeutic option.
- Published
- 2023
- Full Text
- View/download PDF
42. The effect of nafamostat mesilate infusion after ERCP for post-ERCP pancreatitis.
- Author
-
Kim, Joo Seong, Lee, Sang Hyub, Park, Namyoung, Huh, Gunn, Chun, Jung Won, Choi, Jin Ho, Cho, In Rae, Paik, Woo Hyun, Ryu, Ji Kon, and Kim, Yong-Tae
- Subjects
- *
ENDOSCOPIC retrograde cholangiopancreatography , *RETROSPECTIVE studies , *ORGANIC compounds , *PANCREATITIS - Abstract
Background: Nafamostat mesilate decreases the incidence of pancreatitis after endoscopic retrograde cholangiopancreatography (ERCP). However, no studies have administered nafamostat mesilate after ERCP. So we investigated if the infusion of nafamostat mesilate after ERCP can affect the post-ERCP pancreatitis (PEP) in high-risk patients.Methods: In a tertiary hospital, 350 high-risk patients of PEP were reviewed retrospectively. Among them, 201 patients received nafamostat mesilate after ERCP. Patient-related and procedure-related risk factors for PEP were collected. We performed a propensity score matching to adjust for the significant different baseline characteristics. The incidence and severity of PEP were evaluated according to the infusion of nafamostat mesilate. The risk factors of PEP were also analyzed by multivariate logistic regression.Results: The baseline characteristics were not different after the matching. The PEP rate (17.4% vs. 10.3%, P = 0.141) was insignificant. Among the PEP patients, mild PEP was significantly higher in the nafamostat mesilate group (85.7% vs. 45.5%, P = 0.021). Only one patient in the nafamostat mesilate group developed severe PEP. Although young age (odds ratio [OR] 3.60, 95% CI 1.09-11.85, P = 0.035) was a risk factor, nafamostat mesilate (odds ratio [OR] 0.30, 95% CI 0.09-0.98, P = 0.047) was a protective factor for moderate to severe PEP.Conclusions: The administration of nafamostat mesilate after ERCP in high-risk patients was not effective in preventing PEP, but may attenuate the severity of PEP. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
43. Pharmacokinetics of Nafamostat, a Potent Serine Protease Inhibitor, by a Novel LC-MS/MS Analysis.
- Author
-
Oh, Hyeon Seok, Kim, Taehyung, Gu, Dong-Hyeon, Lee, Tae Suk, Kim, Tae Hwan, Shin, Soyoung, and Shin, Beom Soo
- Subjects
- *
LIQUID chromatography-mass spectrometry , *PROTEASE inhibitors , *COVID-19 , *SOLID phase extraction , *CORONAVIRUS disease treatment , *SERINE , *PHARMACOKINETICS - Abstract
Nafamostat, a synthetic serine protease inhibitor, has been used for the treatment of inflammatory diseases such as pancreatitis. Recently, an increasing number of studies have shown the promising antiviral effects of nafamostat for the treatment of coronavirus disease-19 (COVID-19). This study aimed to develop a novel liquid chromatography–tandem mass spectrometry (LC-MS/MS) analysis and to characterize the pharmacokinetics of nafamostat in rats. Nafamostat in the rat plasma was extracted by solid phase extraction, and 13C6-nafamostat was used as an internal standard. The quantification limit of nafamostat in the rat plasma was 0.5 ng/mL. The LC-MS/MS method was fully validated and applied to characterize the pharmacokinetics of nafamostat in rats. Following intravenous injection (2 mg/kg), nafamostat in the plasma showed a multiexponential decline with an average elimination half-life (t1/2) of 1.39 h. Following oral administration of nafamostat solutions (20 mg/kg) in 10% dimethyl sulfoxide (DMSO) and in 10% DMSO with 10% Tween 80, nafamostat was rapidly absorbed, and the average oral bioavailability was 0.95% and 1.59%, respectively. The LC-MS/MS method and the pharmacokinetic information of nafamostat could be helpful for the further preclinical and clinical studies of nafamostat. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
44. Coagulopathy and Fibrinolytic Pathophysiology in COVID-19 and SARS-CoV-2 Vaccination.
- Author
-
Yamada, Shinya and Asakura, Hidesaku
- Subjects
- *
SARS-CoV-2 , *COVID-19 , *DISSEMINATED intravascular coagulation , *COVID-19 vaccines , *AMNIOTIC fluid embolism , *PATHOLOGICAL physiology - Abstract
Coronavirus Disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is frequently complicated by thrombosis. In some cases of severe COVID-19, fibrinolysis may be markedly enhanced within a few days, resulting in fatal bleeding. In the treatment of COVID-19, attention should be paid to both coagulation activation and fibrinolytic activation. Various thromboses are known to occur after vaccination with SARS-CoV-2 vaccines. Vaccine-induced immune thrombotic thrombocytopenia (VITT) can occur after adenovirus-vectored vaccination, and is characterized by the detection of anti-platelet factor 4 antibodies by enzyme-linked immunosorbent assay and thrombosis in unusual locations such as cerebral venous sinuses and visceral veins. Treatment comprises high-dose immunoglobulin, argatroban, and fondaparinux. Some VITT cases show marked decreases in fibrinogen and platelets and marked increases in D-dimer, suggesting the presence of enhanced-fibrinolytic-type disseminated intravascular coagulation with a high risk of bleeding. In the treatment of VITT, evaluation of both coagulation activation and fibrinolytic activation is important, adjusting treatments accordingly to improve outcomes. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
45. Physiologically-based pharmacokinetic modeling of nafamostat to support dose selection for treatment of pediatric patients with COVID-19.
- Author
-
Yong-Soon Cho and Jae-Gook Shin
- Subjects
- *
COVID-19 , *CHILD patients , *MULTISYSTEM inflammatory syndrome , *PEDIATRIC therapy , *PATIENT selection , *AGE groups , *PHARMACOKINETICS - Abstract
Pediatric patients with coronavirus disease 2019 (COVID-19) are increasing, and severe cases such as multisystem inflammatory syndrome are being reported. Nafamostat, a repurposing drug, is currently being explored for the treatment of COVID-19 in adults. However, the data supporting its exposure in pediatrics remains scarce. Physiologically-based pharmacokinetic (PBPK) modeling enables the prediction of drug exposure in pediatrics based on ontogeny of metabolic enzymes and age dependent anatomical and physiological changes. The study aimed to establish a PBPK model of nafamostat in adults, then scale the adult PBPK model to children for predicting pediatric exposures of nafamostat and an optimal weight-based nafamostat dose in pediatric population. The developed model adequately described adult exposure data in healthy volunteers following i.v. administration with three doses (10, 20, and 40 mg). Scaling adult PBPK models to five pediatric groups predicted that as age advances from neonate to adult, the exposure of nafamostat slightly increased from neonate to infant, steadily decreased from infant to child, and then increased from child to adult after the administration of 0.2 mg/kg/h for 14 days, a dosing regimen being conducted in a clinical trial for COVID-19. Based on the fold change of predicted area under the curve for the respective pediatric group over those of adults, weight-based dosages for each pediatric group may be suggested. The novel PBPK model described in this study may be useful to investigate nafamostat pharmacokinetics in a pediatric subgroup further. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
46. Coagulation factors directly cleave SARS-CoV-2 spike and enhance viral entry
- Author
-
Edward R Kastenhuber, Marisa Mercadante, Benjamin Nilsson-Payant, Jared L Johnson, Javier A Jaimes, Frauke Muecksch, Yiska Weisblum, Yaron Bram, Vasuretha Chandar, Gary R Whittaker, Benjamin R tenOever, Robert E Schwartz, and Lewis Cantley
- Subjects
SARS-CoV-2 ,coronavirus ,coagulopathy ,factor Xa ,anticoagulants ,nafamostat ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Coagulopathy is a significant aspect of morbidity in COVID-19 patients. The clotting cascade is propagated by a series of proteases, including factor Xa and thrombin. While certain host proteases, including TMPRSS2 and furin, are known to be important for cleavage activation of SARS-CoV-2 spike to promote viral entry in the respiratory tract, other proteases may also contribute. Using biochemical and cell-based assays, we demonstrate that factor Xa and thrombin can also directly cleave SARS-CoV-2 spike, enhancing infection at the stage of viral entry. Coagulation factors increased SARS-CoV-2 infection in human lung organoids. A drug-repurposing screen identified a subset of protease inhibitors that promiscuously inhibited spike cleavage by both transmembrane serine proteases and coagulation factors. The mechanism of the protease inhibitors nafamostat and camostat may extend beyond inhibition of TMPRSS2 to coagulation-induced spike cleavage. Anticoagulation is critical in the management of COVID-19, and early intervention could provide collateral benefit by suppressing SARS-CoV-2 viral entry. We propose a model of positive feedback whereby infection-induced hypercoagulation exacerbates SARS-CoV-2 infectivity.
- Published
- 2022
- Full Text
- View/download PDF
47. Broad antiviral and anti-inflammatory efficacy of nafamostat against SARS-CoV-2 and seasonal coronaviruses in primary human bronchiolar epithelia.
- Author
-
Niemeyer, Brian F., Miller, Caitlin M., Ledesma-Feliciano, Carmen, Morrison, James H., Jimenez-Valdes, Rocio, Clifton, Clarissa, Poeschla, Eric M., and Benam, Kambez H.
- Subjects
COVID-19 ,SARS-CoV-2 ,CORONAVIRUSES ,EPITHELIUM - Published
- 2022
- Full Text
- View/download PDF
48. Nafamostat reduces systemic inflammation in TLR7-mediated virus-like illness.
- Author
-
Yates, Abi G., Weglinski, Caroline M., Ying, Yuxin, Dunstan, Isobel K., Strekalova, Tatyana, and Anthony, Daniel C.
- Subjects
- *
VIRUS diseases , *IMMUNOMODULATORS , *COVID-19 treatment , *INFLAMMATION , *INTRAPERITONEAL injections - Abstract
Background: The serine protease inhibitor nafamostat has been proposed as a treatment for COVID-19, by inhibiting TMPRSS2-mediated viral cell entry. Nafamostat has been shown to have other, immunomodulatory effects, which may be beneficial for treatment, however animal models of ssRNA virus infection are lacking. In this study, we examined the potential of the dual TLR7/8 agonist R848 to mimic the host response to an ssRNA virus infection and the associated behavioural response. In addition, we evaluated the anti-inflammatory effects of nafamostat in this model.Methods: CD-1 mice received an intraperitoneal injection of R848 (200 μg, prepared in DMSO, diluted 1:10 in saline) or diluted DMSO alone, and an intravenous injection of either nafamostat (100 μL, 3 mg/kg in 5% dextrose) or 5% dextrose alone. Sickness behaviour was determined by temperature, food intake, sucrose preference test, open field and forced swim test. Blood and fresh liver, lung and brain were collected 6 h post-challenge to measure markers of peripheral and central inflammation by blood analysis, immunohistochemistry and qPCR.Results: R848 induced a robust inflammatory response, as evidenced by increased expression of TNF, IFN-γ, CXCL1 and CXCL10 in the liver, lung and brain, as well as a sickness behaviour phenotype. Exogenous administration of nafamostat suppressed the hepatic inflammatory response, significantly reducing TNF and IFN-γ expression, but had no effect on lung or brain cytokine production. R848 administration depleted circulating leukocytes, which was restored by nafamostat treatment.Conclusions: Our data indicate that R848 administration provides a useful model of ssRNA virus infection, which induces inflammation in the periphery and CNS, and virus infection-like illness. In turn, we show that nafamostat has a systemic anti-inflammatory effect in the presence of the TLR7/8 agonist. Therefore, the results indicate that nafamostat has anti-inflammatory actions, beyond its ability to inhibit TMPRSS2, that might potentiate its anti-viral actions in pathologies such as COVID-19. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
49. Retroperitoneal hemorrhage in a patient with coronavirus disease 2019 (COVID-19) : A case report.
- Author
-
Takuya Yoshioka, Kei Daizumoto, Kouki Tada, Masato Mima, Kozo Kagawa, Tomoya Fukawa, Kunihisa Yamaguchi, Masayuki Takahashi, Yasuhiko Nishioka, and Hiroomi Kanayama
- Subjects
HEMORRHAGE treatment ,RETROPERITONEUM diseases ,ANTICOAGULANTS ,HEMATOLOGIC agents ,BLOOD coagulation - Abstract
Introduction: Early prophylactic administration of anticoagulants is recommended in patients with coronavirus disease 2019 (COVID-19). A case of retroperitoneal hemorrhage during inpatient treatment for COVID-19 is reported. Case Presentation : A 69-year-old man was diagnosed with COVID-19 6 days after symptom onset. After admission for difficulty of breathing, he was treated with steroid pulse therapy, remdesivir, and heparin sodium. On day 16 after admission, his hemoglobin and blood pressure dropped. Computed tomography showed a left retroperitoneal hematoma and multiple areas of extravasation in bilateral iliopsoas muscles. Anticoagulation therapy was stopped, and blood transfusion therapy was chosen by considering poor general condition caused by severe pneumonia. On day 19, the hemoglobin and blood pressure improved, and blood transfusion was stopped. However, he died on day 25 due to pneumonia. Conclusion : When retroperitoneal hemorrhage occurs as a complication of COVID-19, appropriate treatment decision, transcatheter arterial embolization or conservative treatment, should be chosen based on patient's condition. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
50. Physical Compatibility of Nafamostat with Analgesics, Sedatives, and Muscle Relaxants for Treatment of Coronavirus Disease 2019.
- Author
-
Masayoshi Kondo, Makihiko Nagano, Mariko Yoshida, Naoki Yoshida, Naoya Tagui, Masato Yoshida, Kazutoshi Sugaya, and Hisamitsu Takase
- Subjects
- *
CORONAVIRUS disease treatment , *COVID-19 , *MUSCLE relaxants , *ANALGESICS , *SEDATIVES , *ROCURONIUM bromide - Abstract
Background: Severe coronavirus disease 2019 (COVID-19) may require continuous administration of analgesics, sedatives, and muscle relaxants. Nafamostat has recently been reported as a therapeutic agent for COVID-19. However, there is a lack of information on the compatibility of nafamostat with the aforementioned drug classes. This study evaluated the physical compatibility of nafamostat with these drug classes. Methods: Nafamostat was combined with 1-3 target drugs (fentanyl, morphine, midazolam, dexmedetomidine, and rocuronium). Fifteen physical compatibility tests were conducted. Nafamostat was dissolved in 5% glucose solution; the final concentration was 10 mg/mL. All other medications were diluted in 0.9% sodium chloride to obtain clinically relevant concentrations. The power of hydrogen (pH) of all medications was measured during each test. Compatibility tests were conducted with 4 test solutions in which nafamostat and the target drugs were compounded at equal volume ratios (1:1, 1:1:1, or 1:1:1:1). Visual appearance, turbidity, and pH were evaluated immediately after mixing and at 1 and 3 hours. Physical incompatibilities were defined as gross precipitation, cloudiness, appearance of the Tyndall effect, or a turbidity change of ≥0.5 nephelometric turbidity units (NTU) based on nafamostat. Results: The mean pH of nafamostat was 3.13 ± 0.03. The combination of nafamostat, fentanyl, and dexmedetomidine had the highest pH (3.39 ± 0.01; 3 hours after mixing). All drugs were compatible with nafamostat until 3 hours after admixture, with a mean turbidity value of ≤0.03 NTU. Conclusions: Infusions combining nafamostat with the tested sedatives, analgesics, and muscle relaxants could be safely administered. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.