Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 174
Filter
1.
Clin Pharmacol Ther ; 115(2): 206-212, 2024 02.
Article in English | MEDLINE | ID: mdl-38032816

ABSTRACT

Pancreatitis is the most common complication of endoscopic retrograde cholangiopancreatography (ERCP). As the management of pancreatitis is limited, clinical approaches focus on the prevention of post-ERCP pancreatitis (PEP). In theory, the serine protease inhibitor nafamostat can reduce circulating inflammatory mediators in pancreatitis. We aimed to investigate the effect of nafamostat in the prevention of PEP in this systematic review and meta-analysis. The protocol for this review was registered in PROSPERO (CRD42022367988). We systematically searched 5 databases without any filters on September 26, 2022. The eligible population was adult patients undergoing ERCP. We compared the PEP preventive effect of nafamostat to placebo. The main outcome was the occurrence of PEP. We calculated the pooled odds ratios (ORs), mean differences, and corresponding 95% confidence intervals (95% CIs) and multilevel model. The risk of bias was assessed using the Rob2 tool. Seven randomized controlled trials involving 2,962 patients were eligible for inclusion. Nafamostat reduced the overall incidence rate of PEP (20 mg, OR: 0.50, 95% CI: 0.30-0.82 and 50 mg, OR: 0.48, 95% CI: 0.24-0.96). However, the occurrence of mild PEP was significantly reduced only in the subgroup receiving 20 mg nafamostat (OR, 0.49, 95% CI: 0.31-0.77). Overall, nafamostat therapy reduced moderate PEP in high-risk patients (OR: 0.18, 95% CI: 0.0.4-0.84) and mild PEP in low-risk patients (OR: 0.32, 95% CI: 0.17-0.61). Nafamostat is an effective therapy in the prevention of mild post-ERCP pancreatitis. Further research is required to determine the cost-effectiveness of this therapy.


Subject(s)
Benzamidines , Cholangiopancreatography, Endoscopic Retrograde , Guanidines , Pancreatitis , Adult , Humans , Benzamidines/therapeutic use , Cholangiopancreatography, Endoscopic Retrograde/adverse effects , Guanidines/therapeutic use , Incidence , Pancreatitis/epidemiology , Pancreatitis/etiology , Pancreatitis/prevention & control , Randomized Controlled Trials as Topic
2.
J Burn Care Res ; 45(3): 764-770, 2024 05 06.
Article in English | MEDLINE | ID: mdl-38113522

ABSTRACT

Continuous renal replacement therapy (CRRT) is often disrupted due to various factors, such as patient-related issues, vascular access complications, treatment plans, and medical staff factors. This unexpected interruption is referred to as non-selective filter stoppage and can result in additional treatment expenses. This study conducted a retrospectively analyzed 501 CRRT filters used in 62 patients with severe burns, lifespan and therapeutic effect of all filters were mainly analyzed, used logistic regression analysis was performed to identify risk factors associated with non-selective cessation filters. Out of 493 filters, 279 cases received heparin (56.60%), the median lifespan of the filter was 14.08 h (25th, 75th quantile: 7.30, 21.50); 128 cases were treated with nafamostat mesylate (26.00%), and the median lifespan of the filter was 16.42 h (10.49, 22.76); 86 cases were treated with sodium citrate (17.40%), and the median lifespan of the filter was 31.06 h (19.25, 48.75). In addition, significant differences were observed in the electrolyte index, renal function index, and procalcitonin levels before and after treatment with a single filter (P < .001). Multivariate logistic regression showed that the risk of non-selective cessation of sodium citrate anticoagulants was lower than that of heparin anticoagulation. Overall, CRRT is progressively becoming more prevalent in the treatment of patients with severe burns. The lifespan of individual filters and total patient treatment duration showed a consistent upward trend. The filter's lifespan was notably greater during sodium citrate anticoagulation when compared to nafamostat mesylate and heparin, meanwhile notably reducing the risk of non-selective cessation. Therefore, we recommend sodium citrate for anticoagulation in patients without any contraindications.


Subject(s)
Burns , Continuous Renal Replacement Therapy , Humans , Burns/therapy , Female , Male , Retrospective Studies , Middle Aged , Adult , Anticoagulants/therapeutic use , Guanidines/therapeutic use , Benzamidines/therapeutic use , Acute Kidney Injury/therapy , Heparin/therapeutic use , Aged , Sodium Citrate/therapeutic use , Treatment Outcome , Risk Factors , Citrates/therapeutic use , Time Factors
3.
Eur J Pharmacol ; 919: 174795, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35122868

ABSTRACT

N-methyl-D-aspartate (NMDA) receptors are affected by many pharmaceuticals. In this work, we studied the action of the serine protease inhibitors nafamostat, gabexate and camostat, and an antiprotozoal compound, furamidine, on native NMDA receptors in rat hippocampal pyramidal neurons. Nafamostat, furamidine and gabexate inhibited these receptors with IC50 values of 0.20 ± 0.04, 0.64 ± 0.13 and 16 ± 3 µM, respectively, whereas camostat was ineffective. Nafamostat and furamidine showed voltage-dependent inhibition, while gabexate showed practically voltage-independent inhibition. Nafamostat and furamidine demonstrated tail currents, implying a 'foot-in-the-door' mechanism of action; gabexate did not demonstrate any signs of 'foot-in-the-door' or trapping channel block. Gabexate action was also not competitive, suggesting allosteric inhibition of NMDA receptors. Furamidine and nafamostat are structurally similar to the previously studied diminazene and all three demonstrated a 'foot-in-the-door' mechanism. They have a rather rigid, elongated structures and cannot fold into more compact forms. By contrast, the gabexate molecule can fold, but its folded structure differs drastically from that of typical NMDA receptor blockers, in agreement with its voltage-independent inhibition. These findings provide a better understanding of the structural determinants of NMDA receptor antagonism, while also supporting the potential clinical repurposing of these drugs as neuroprotectors for glaucoma and other neurodegenerative diseases.


Subject(s)
Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Animals , Benzamidines/pharmacology , Benzamidines/therapeutic use , Drug Repositioning , Esters/pharmacology , Esters/therapeutic use , Gabexate/pharmacology , Gabexate/therapeutic use , Guanidines/pharmacology , Guanidines/therapeutic use , Hippocampus/drug effects , Inhibitory Concentration 50 , Male , Models, Animal , Neurodegenerative Diseases/drug therapy , Pyramidal Cells/drug effects , Rats , Rats, Wistar , Serine Proteinase Inhibitors/therapeutic use
4.
EBioMedicine ; 76: 103856, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35152152

ABSTRACT

BACKGROUND: Many repurposed drugs have progressed rapidly to Phase 2 and 3 trials in COVID19 without characterisation of Pharmacokinetics /Pharmacodynamics including safety data. One such drug is nafamostat mesylate. METHODS: We present the findings of a phase Ib/IIa open label, platform randomised controlled trial of intravenous nafamostat in hospitalised patients with confirmed COVID-19 pneumonitis. Patients were assigned randomly to standard of care (SoC), nafamostat or an alternative therapy. Nafamostat was administered as an intravenous infusion at a dose of 0.2 mg/kg/h for a maximum of seven days. The analysis population included those who received any dose of the trial drug and all patients randomised to SoC. The primary outcomes of our trial were the safety and tolerability of intravenous nafamostat as an add on therapy for patients hospitalised with COVID-19 pneumonitis. FINDINGS: Data is reported from 42 patients, 21 of which were randomly assigned to receive intravenous nafamostat. 86% of nafamostat-treated patients experienced at least one AE compared to 57% of the SoC group. The nafamostat group were significantly more likely to experience at least one AE (posterior mean odds ratio 5.17, 95% credible interval (CI) 1.10 - 26.05) and developed significantly higher plasma creatinine levels (posterior mean difference 10.57 micromol/L, 95% CI 2.43-18.92). An average longer hospital stay was observed in nafamostat patients, alongside a lower rate of oxygen free days (rate ratio 0.55-95% CI 0.31-0.99, respectively). There were no other statistically significant differences in endpoints between nafamostat and SoC. PK data demonstrated that intravenous nafamostat was rapidly broken down to inactive metabolites. We observed no significant anticoagulant effects in thromboelastometry. INTERPRETATION: In hospitalised patients with COVID-19, we did not observe evidence of anti-inflammatory, anticoagulant or antiviral activity with intravenous nafamostat, and there were additional adverse events. FUNDING: DEFINE was funded by LifeArc (an independent medical research charity) under the STOPCOVID award to the University of Edinburgh. We also thank the Oxford University COVID-19 Research Response Fund (BRD00230).


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Benzamidines/therapeutic use , COVID-19 Drug Treatment , Guanidines/therapeutic use , Administration, Intravenous , Adult , Aged , Aged, 80 and over , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Benzamidines/adverse effects , Benzamidines/pharmacokinetics , Biomarkers/blood , Biomarkers/metabolism , COVID-19/mortality , COVID-19/virology , Drug Administration Schedule , Female , Guanidines/adverse effects , Guanidines/pharmacokinetics , Half-Life , Humans , Immunophenotyping , Kaplan-Meier Estimate , Male , Middle Aged , SARS-CoV-2/isolation & purification , SARS-CoV-2/physiology , Treatment Outcome , Viral Load
5.
J Neuroinflammation ; 19(1): 8, 2022 Jan 06.
Article in English | MEDLINE | ID: mdl-34991643

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.


Subject(s)
Benzamidines , Guanidines , Inflammation/drug therapy , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors , Toll-Like Receptor 7/immunology , Virus Diseases/drug therapy , Animals , Benzamidines/pharmacology , Benzamidines/therapeutic use , COVID-19/complications , Guanidines/pharmacology , Guanidines/therapeutic use , Illness Behavior/drug effects , Imidazoles/administration & dosage , Imidazoles/immunology , Inflammation/metabolism , Inflammation/virology , Male , Mice , Serine Proteinase Inhibitors/pharmacology , Serine Proteinase Inhibitors/therapeutic use , Toll-Like Receptor 7/agonists , Virus Diseases/metabolism , Virus Diseases/virology , COVID-19 Drug Treatment
6.
Viruses ; 13(9)2021 09 04.
Article in English | MEDLINE | ID: mdl-34578348

ABSTRACT

SARS-CoV-2 and its vaccine/immune-escaping variants continue to pose a serious threat to public health due to a paucity of effective, rapidly deployable, and widely available treatments. Here, we address these challenges by combining Pegasys (IFNα) and nafamostat to effectively suppress SARS-CoV-2 infection in cell culture and hamsters. Our results indicate that Serpin E1 is an important mediator of the antiviral activity of IFNα and that both Serpin E1 and nafamostat can target the same cellular factor TMPRSS2, which plays a critical role in viral replication. The low doses of the drugs in combination may have several clinical advantages, including fewer adverse events and improved patient outcome. Thus, our study may provide a proactive solution for the ongoing pandemic and potential future coronavirus outbreaks, which is still urgently required in many parts of the world.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzamidines/pharmacology , COVID-19/metabolism , COVID-19/virology , Guanidines/pharmacology , Interferon-alpha/pharmacology , SARS-CoV-2/drug effects , Serine Endopeptidases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Benzamidines/therapeutic use , Cricetinae , Disease Models, Animal , Drug Therapy, Combination , Female , Guanidines/therapeutic use , Host-Pathogen Interactions/drug effects , Humans , Interferon-alpha/therapeutic use , Virus Replication/drug effects , COVID-19 Drug Treatment
8.
Int J Mol Sci ; 22(6)2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33802169

ABSTRACT

In order to treat Coronavirus Disease 2019 (COVID-19), we predicted and implemented a drug delivery system (DDS) that can provide stable drug delivery through a computational approach including a clustering algorithm and the Schrödinger software. Six carrier candidates were derived by the proposed method that could find molecules meeting the predefined conditions using the molecular structure and its functional group positional information. Then, just one compound named glycyrrhizin was selected as a candidate for drug delivery through the Schrödinger software. Using glycyrrhizin, nafamostat mesilate (NM), which is known for its efficacy, was converted into micelle nanoparticles (NPs) to improve drug stability and to effectively treat COVID-19. The spherical particle morphology was confirmed by transmission electron microscopy (TEM), and the particle size and stability of 300-400 nm were evaluated by measuring DLSand the zeta potential. The loading of NM was confirmed to be more than 90% efficient using the UV spectrum.


Subject(s)
COVID-19 Drug Treatment , Computational Biology/methods , Drug Delivery Systems/methods , A549 Cells , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Benzamidines/chemistry , Benzamidines/therapeutic use , Cell Survival/drug effects , Cluster Analysis , Computer Simulation , Databases, Pharmaceutical , Drug Carriers/chemistry , Drug Repositioning , Drug Stability , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/therapeutic use , Guanidines/chemistry , Guanidines/therapeutic use , Humans , Hydrophobic and Hydrophilic Interactions , Micelles , Microscopy, Electron, Transmission , Molecular Structure , Nanoparticles/chemistry , Particle Size
9.
J Nippon Med Sch ; 88(6): 533-539, 2021 Dec 29.
Article in English | MEDLINE | ID: mdl-33692301

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.


Subject(s)
Analgesics/therapeutic use , Benzamidines/therapeutic use , COVID-19 Drug Treatment , Drug Incompatibility , Fentanyl/therapeutic use , Guanidines/therapeutic use , Muscle Relaxants, Central/therapeutic use , Dexmedetomidine/therapeutic use , Humans , Hypnotics and Sedatives , SARS-CoV-2 , Treatment Outcome
10.
Appl Biochem Biotechnol ; 193(6): 1909-1923, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33512650

ABSTRACT

The unprecedented coronavirus SARS-CoV-2 outbreak at Wuhan, China, caused acute respiratory infection to humans. There is no precise vaccine/therapeutic agents available to combat the COVID-19 disease. Some repurposed drugs are saving the life of diseased, but the complete cure is relatively less. Several drug targets have been reported to inhibit the SARS-CoV-2 virus infection, in that TMPRSS2 (transmembrane protease serine 2) is one of the potential targets; inhibiting this protease stops the virus entry into the host human cell. Camostat mesylate, nafamostat, and leupeptin are the drugs, in which the first two drugs are being used for COVID-19 and leupeptin also tested. To consider these drugs as the repurposed drug for COVID-19, it is essential to understand their binding affinity and stability with TMPRSS2. In the present study, we performed the molecular docking and molecular dynamics (MD) simulation of these molecules with the TMPRSS2. The docking study reveals that leupeptin molecule strongly binds with TMPRSS2 protein than the other two drug molecules. The RMSD and RMSF values of MD simulation confirm that leupeptin and the amino acids of TMPRSS2 are very stable than the other two molecules. Furthermore, leupeptin forms interactions with the key amino acids of TMPRSS2 and the same have been maintained during the MD simulations. This structural and dynamical information is useful to evaluate these drugs to be used as repurposed drugs, however, the strong binding profile of leupeptin with TMPRSS2, suggests, it may be considered as a repurposed drug for COVID-19 disease after clinical trial.


Subject(s)
Antiviral Agents/pharmacology , Benzamidines/therapeutic use , COVID-19 Drug Treatment , Drug Repositioning , Esters/therapeutic use , Guanidines/therapeutic use , Leupeptins/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Serine Endopeptidases/metabolism , Antiviral Agents/therapeutic use , Benzamidines/pharmacology , COVID-19/virology , Esters/pharmacology , Guanidines/pharmacology , Humans , Protein Binding , SARS-CoV-2/drug effects
11.
Dig Dis Sci ; 66(12): 4475-4484, 2021 12.
Article in English | MEDLINE | ID: mdl-33495919

ABSTRACT

BACKGROUND: Endoscopic retrograde cholangiopancreatography (ERCP) is associated with complications such as post-ERCP pancreatitis (PEP). Protease inhibitors, including nafamostat mesylate (NM), have been evaluated for prophylaxis against PEP. AIM: We describe the first multicenter randomized controlled trial assessing the prophylactic efficacy of NM against PEP. METHODS: In this multicenter prospective study, we aimed to enroll 800 patients aged ≥ 20 years with a planned ERCP between December 2012 and March 2019. The primary outcome was the incidence and severity of PEP in patients who did not receive NM (non-NM) versus those who did (NM; 20 mg). Secondary outcomes included the incidence of PEP by NM initiation (pre- and post-ERCP), risk factors for PEP, and NM-related adverse events. RESULTS: Only 441 of the planned 800 patients were enrolled (non-NM: n = 149; NM: n = 292 [pre-ERCP NM: n = 144; post-ERCP NM: n = 148]). Patient characteristics were balanced at baseline with no significant differences between groups. PEP occurred in 40/441 (9%) patients (non-NM: n = 15 [10%]; NM: n = 25 [9%]), including 17 (12%) and eight (8%) in the pre-ERCP and post-ERCP NM groups, respectively. In the NM group, the incidence of PEP was lower in the low-risk group than in the high-risk group. Pancreatic injection and double-guidewire technique were independent risk factors for PEP. NM-related adverse events of hyperkalemia occurred in two (0.7%) patients. CONCLUSIONS: We found no evidence for the prophylactic effect of NM against PEP, regardless of the timing of administration; however, further studies are needed.


Subject(s)
Benzamidines/therapeutic use , Cholangiopancreatography, Endoscopic Retrograde/adverse effects , Guanidines/therapeutic use , Pancreatitis/prevention & control , Trypsin Inhibitors/therapeutic use , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Pancreatitis/etiology , Prospective Studies
12.
Cornea ; 40(6): 769-773, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-32833843

ABSTRACT

PURPOSE: To report endotheliitis as an early, key presenting sign of Acanthamoeba keratitis in patients who are soft contact lens wearers. METHODS: We report 4 cases of patients presenting with pain, red eye, and reduction in vision in the context of soft contact lens wear. On examination, the predominant clinical finding was that of endotheliitis, localized fusiform stromal edema with corresponding keratic precipitates, anterior chamber inflammation, and minimal epithelial and anterior stromal signs. The classical signs of Acanthamoeba keratitis were not present. RESULTS: All 4 cases were clinically diagnosed as Acanthamoeba endotheliitis; corneal scrapes were negative; case 1 was polymerase chain reaction positive, and case 3 underwent confocal microscopy that showed double-walled cysts, suggesting Acanthamoeba. All responded well to anti-Acanthamoeba medication alone with 3 cases achieving complete resolution with minimal anterior stromal scarring by 7 weeks. Case 1 had steroid treatment before being seen at our unit and had a prolonged course of treatment with complete resolution by 4 months. CONCLUSIONS: It is imperative to have a high index of suspicion for Acanthamoeba in patients presenting with pain and endotheliitis in the context of contact lens wear, even in the absence of classical signs.


Subject(s)
Acanthamoeba Keratitis/diagnosis , Endothelium, Corneal/pathology , Inflammation/diagnosis , Acanthamoeba Keratitis/drug therapy , Acanthamoeba Keratitis/etiology , Administration, Ophthalmic , Adult , Antiprotozoal Agents/therapeutic use , Benzamidines/therapeutic use , Biguanides/therapeutic use , Contact Lenses, Hydrophilic/adverse effects , Disinfectants/therapeutic use , Female , Humans , Inflammation/drug therapy , Inflammation/etiology , Male , Microscopy, Confocal , Middle Aged , Ophthalmic Solutions , Polymerase Chain Reaction
13.
J Thromb Thrombolysis ; 51(3): 649-656, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32920751

ABSTRACT

Critical illnesses associated with coronavirus disease 2019 (COVID-19) are attributable to a hypercoagulable status. There is limited knowledge regarding the dynamic changes in coagulation factors among COVID-19 patients on nafamostat mesylate, a potential therapeutic anticoagulant for COVID-19. First, we retrospectively conducted a cluster analysis based on clinical characteristics on admission to identify latent subgroups among fifteen patients with COVID-19 on nafamostat mesylate at the University of Tokyo Hospital, Japan, between April 6 and May 31, 2020. Next, we delineated the characteristics of all patients as well as COVID-19-patient subgroups and compared dynamic changes in coagulation factors among each subgroup. The subsequent dynamic changes in fibrinogen and D-dimer levels were presented graphically. All COVID-19 patients were classified into three subgroups: clusters A, B, and C, representing low, intermediate, and high risk of poor outcomes, respectively. All patients were alive 30 days from symptom onset. No patient in cluster A required mechanical ventilation; however, all patients in cluster C required mechanical ventilation, and half of them were treated with venovenous extracorporeal membrane oxygenation. All patients in cluster A maintained low D-dimer levels, but some critical patients in clusters B and C showed dynamic changes in fibrinogen and D-dimer levels. Although the potential of nafamostat mesylate needs to be evaluated in randomized clinical trials, admission characteristics of patients with COVID-19 could predict subsequent coagulopathy.


Subject(s)
Anticoagulants/therapeutic use , Benzamidines/therapeutic use , COVID-19 Drug Treatment , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinogen/metabolism , Guanidines/therapeutic use , Aged , Anticoagulants/pharmacology , Benzamidines/pharmacology , COVID-19/blood , COVID-19/classification , Female , Fibrinogen/drug effects , Guanidines/pharmacology , Humans , Male , Middle Aged , Retrospective Studies
14.
J Med Virol ; 93(6): 3484-3495, 2021 06.
Article in English | MEDLINE | ID: mdl-33247612

ABSTRACT

The effects of the clinically used protease inhibitor nafamostat on influenza virus replication have not been well studied. Primary human tracheal (HTE) and nasal (HNE) epithelial cells were pretreated with nafamostat and infected with the 2009 pandemic [A/Sendai-H/108/2009/(H1N1) pdm09] or seasonal [A/New York/55/2004(H3N2)] influenza virus. Pretreatment with nafamostat reduced the titers of the pandemic and seasonal influenza viruses and the secretion of inflammatory cytokines, including interleukin-6 and tumor necrosis factor-α, in the supernatants of the cells infected with the pandemic influenza virus. HTE and HNE cells exhibited mRNA and/or protein expression of transmembrane protease serine 2 (TMPRSS2), TMPRSS4, and TMPRSS11D. Pretreatment with nafamostat reduced cleavage of the precursor protein HA0 of the pandemic influenza virus into subunit HA1 in HTE cells and reduced the number of acidic endosomes in HTE and HNE cells where influenza virus RNA enters the cytoplasm. Additionally, nafamostat (30 mg/kg/day, intraperitoneal administration) reduced the levels of the pandemic influenza virus [A/Hyogo/YS/2011 (H1N1) pdm09] in mouse lung washes. These findings suggest that nafamostat may inhibit influenza virus replication in human airway epithelial cells and mouse lungs and reduce infection-induced airway inflammation by modulating cytokine production.


Subject(s)
Benzamidines/pharmacology , Benzamidines/therapeutic use , Epithelial Cells/drug effects , Guanidines/pharmacology , Guanidines/therapeutic use , Lung/drug effects , Serine Proteinase Inhibitors/pharmacology , Serine Proteinase Inhibitors/therapeutic use , Virus Replication/drug effects , Animals , Cells, Cultured , Cytokines/analysis , Cytokines/immunology , Epithelial Cells/virology , Female , Humans , Lung/virology , Male , Mice , Mice, Inbred BALB C , Nose/cytology , Trachea/cytology
15.
Curr Eye Res ; 46(6): 771-776, 2021 06.
Article in English | MEDLINE | ID: mdl-33151784

ABSTRACT

Purpose: To unveil the long-term prognosis of Acanthamoeba keratitis based on clinical presentation and timing of diagnosis to better inform patients since the first visit regarding their length of treatment, quality of life, and visual function.Methods: Retrospective observational study enrolling patients with Acanthamoeba keratitis from 1994 to 2019. Patients with a complete eye examination and medical records were analyzed. The severity of the disease, the time from onset of symptoms to the appropriate therapeutic regimen, the time until clinical resolution, visual function, and long term follow-up was evaluated. Quality of life was assessed at the last follow-up visit by means of the VFQ-25 questionnaire.Results: Thirty-five patients (40 eyes) were assessed. The overall healing time of patients with Acanthamoeba keratitis was 12.5 ± 3.5 months, while patients with a severe corneal ulcer (stage III) had a significant longer healing time (16.2 ± 3.7 months) compared to patients with stage II (7.04 ± 0.7 months) or I (7.7 ± 1.5 months; p < .05). Patients who received a prompt therapy (<30 days form symptoms onset) had a reduced healing time compared to patients with a delayed diagnosis (p < .01). Quality of life was assessed after a mean of 11.7 ± 4.7 years and it was mildly reduced (86.6 ± 17). Patients that were diagnosed early (<30 days from onset) showed a lower reduction in quality of life than in patients that were diagnosed >30 days from onset. After resolution, 59% of the patients considered unnecessary any further proposed surgical intervention.Conclusions: Delayed diagnosis of Acanthamoeba keratitis and disease severity significantly increases healing time and duration of treatment. The time to diagnosis and disease stage at diagnosis predicts the duration of treatment, the final outcome, quality of life, and the requirement of surgery. These data would allow us to promptly inform patients about long-term disease timeline, future outcomes, improving disease acceptance, and quality of life.


Subject(s)
Acanthamoeba Keratitis/diagnosis , Acanthamoeba Keratitis/physiopathology , Acanthamoeba Keratitis/drug therapy , Acanthamoeba Keratitis/psychology , Adolescent , Adult , Aged , Antiprotozoal Agents/therapeutic use , Benzamidines/therapeutic use , Biguanides/therapeutic use , Child , Contact Lenses/adverse effects , Corneal Ulcer/physiopathology , Corneal Ulcer/psychology , Disinfectants/therapeutic use , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Prognosis , Quality of Life/psychology , Retrospective Studies , Risk Factors , Time Factors , Visual Acuity/physiology , Wound Healing/physiology , Young Adult
16.
Middle East Afr J Ophthalmol ; 27(4): 244-246, 2020.
Article in English | MEDLINE | ID: mdl-33814825

ABSTRACT

We are reporting the case of a 25-year-old female who developed acanthamoeba keratitis after wearing contact lenses for high myopia. She was diagnosed as acanthamoeba and started the treatment of antiacanthamoeba for 3 consecutive weeks, followed by bare Descemet's therapeutic lamellar keratoplasty (LKP) with the maintenance of antiacanthamoeba treatment to control the infection. In the late postoperative period, visual rehabilitation was obtained by insertion of implantable Collamer lens (ICL) with her final visual outcome was 20/30. For acanthamoeba keratitis, early bare Descemet's therapeutic LKP has a better outcome in comparison to late penetrating keratoplasty in terms of infection eradication and globe preservation. After removal of all sutures, the refractive error can be corrected with photorefractive procedures as well as ICL.


Subject(s)
Acanthamoeba Keratitis/therapy , Anti-Infective Agents, Local/therapeutic use , Antifungal Agents/therapeutic use , Corneal Transplantation , Lenses, Intraocular , Vision Disorders/rehabilitation , Acanthamoeba Keratitis/drug therapy , Acanthamoeba Keratitis/surgery , Adult , Benzamidines/therapeutic use , Chlorhexidine/therapeutic use , Drug Therapy, Combination , Female , Fluconazole/therapeutic use , Humans , Slit Lamp Microscopy , Visual Acuity/physiology
17.
Anticancer Agents Med Chem ; 20(4): 450-463, 2020.
Article in English | MEDLINE | ID: mdl-31736450

ABSTRACT

BACKGROUND: Colorectal cancer is among the leading causes of death worldwide. The incidence of deaths is expected to be 11.4 million in 2030. OBJECTIVE: We aimed to evaluate the in vitro and in vivo antioxidant and antitumor activities of a novel Bithiophene- Fluorobenzamidine (BFB) against DMH-induced colorectal cancer in rats. METHODS: The antiproliferative activity of BFB against HCT-116 colon cancer cells and apoptotic genes was assessed. In vivo study was also conducted in which 80 adult male rats were divided into 5 groups; control, BFB, and the other 3 groups were injected with DMH (20mg/kg, s.c., for 9 weeks). Group 4 was injected with 5 doses of cisplatin (2.5mg/kg, i.p over 21 weeks) and group 5 was injected with 3 doses/week of BFB (2.5mg/kg, i.p, for 21 weeks). RESULTS: BFB exhibited weak to moderate in vitro antioxidant activity. It had a strong antiproliferative activity with IC50 ~0.3µg/ml. BFB induced extrinsic apoptosis through the upregulation of FasL, TRAL, p53 and caspase-8, and intrinsic apoptosis through the downregulation of Bcl-2 and survivin. BFB decreased the tumor incidence, multiplicity and size and improved the decreased body weight. BFB also ameliorated the functions of kidney and liver and antioxidants deteriorated by DMH. BFB significantly improved the pathological changes caused by DMH in colon tissues. CONCLUSION: BFB showed a very promising antitumor activity against colorectal cancer induced by DMH in rats without causing hepato- or nephrotoxicity.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Benzamidines/chemistry , Benzamidines/therapeutic use , Colorectal Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Antioxidants/therapeutic use , Apoptosis/drug effects , Benzamidines/pharmacology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/pathology , Dimethylhydrazines , Drug Discovery , HCT116 Cells , Halogenation , Humans , Male , Rats
18.
Cont Lens Anterior Eye ; 42(5): 506-511, 2019 10.
Article in English | MEDLINE | ID: mdl-31018907

ABSTRACT

PURPOSE OF THE STUDY: To report an observational study of Acanthamoeba Keratitis (AK) in University Hospital Coventry & Warwickshire (UHCW), Coventry, UK and determine risk factors, outcomes as well as incidence rates. METHODS: A retrospective analysis was done of consecutive patients who were treated for AK by the corneal service at UHCW from January 2017 to January 2018. Cases were identified from 2 sources; the department of microbiology and the hospital pharmacy. Patient data was collected by 3 of the authors using both paper and electronic medical records. Information was also gathered over the telephone using a predefined questionnaire. The primary endpoint used for assessing duration of disease was time to resolution after the initial diagnosis. RESULTS: 9 eyes were identified over the 12-month period, a drastic increase from previous years. All were contact lens (CL) wearers and 3 used daily disposable CL's. 8 out of 9 patients had an improvement in best corrected visual acuity (BCVA) which was consistent with their baseline levels pre-infection. The average length of treatment was 107.25 days for the 8 resolved cases. 1 case is still having ongoing treatment. No case has required surgical treatment. Several patients admitted to exposing their CL to unsterile water either via swimming, showering or tap water. All patients had purchased their lenses from contact lens practitioners. CONCLUSION: Overall, our study shows excellent outcomes. Almost all patients had resolution of symptoms with medical treatment and an improvement in visual acuity. Several contributing factors have aided us in achieving this including early diagnosis, a robust treatment protocol and diagnostic modalities such as Polymer Chain Reaction (PCR) and Confocal Microscopy (CFM). However, the increased incidence compared to previous years is a worrying trend and there will be an ongoing analysis looking at patterns of incidence in the future.


Subject(s)
Acanthamoeba Keratitis/epidemiology , Hospitals, Teaching/statistics & numerical data , Acanthamoeba/genetics , Acanthamoeba Keratitis/diagnosis , Acanthamoeba Keratitis/drug therapy , Adult , Anti-Infective Agents, Local/therapeutic use , Antiprotozoal Agents/therapeutic use , Benzamidines/therapeutic use , Biguanides/therapeutic use , Chlorhexidine/therapeutic use , Contact Lenses/adverse effects , DNA, Protozoan/genetics , Disinfectants/therapeutic use , Female , Humans , Incidence , Male , Middle Aged , Polymerase Chain Reaction , Retrospective Studies , Risk Factors , United Kingdom/epidemiology , Visual Acuity/physiology , Young Adult
19.
ACS Chem Biol ; 13(9): 2708-2718, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30118588

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an autosomal dominant, CTG•CAG microsatellite expansion disease. Expanded CUG repeat RNA sequester the muscleblind-like (MBNL) family of RNA-binding proteins, thereby disrupting their normal cellular function which leads to global mis-regulation of RNA processing. Previously, the small molecule furamidine was shown to reduce CUG foci and rescue mis-splicing in a DM1 HeLa cell model and to rescue mis-splicing in the HSALR DM1 mouse model, but furamidine's mechanism of action was not explored. Here we use a combination of biochemical, cell toxicity, and genomic studies in DM1 patient-derived myotubes and the HSALR DM1 mouse model to investigate furamidine's mechanism of action. Mis-splicing rescue was observed in DM1 myotubes and the HSALR DM1 mouse with furamidine treatment. Interestingly, while furamidine was found to bind CTG•CAG repeat DNA with nanomolar affinity, a reduction in expanded CUG repeat transcript levels was observed in the HSALR DM1 mouse but not DM1 patient-derived myotubes. Further investigation in these cells revealed that furamidine treatment at nanomolar concentrations led to up-regulation of MBNL1 and MBNL2 protein levels and a reduction of ribonuclear foci. Additionally, furamidine was shown to bind CUG RNA with nanomolar affinity and disrupted the MBNL1 -CUG RNA complex in vitro at micromolar concentrations. Furamidine's likely promiscuous interactions in vitro and in vivo appear to affect multiple pathways in the DM1 mechanism to rescue mis-splicing, yet surprisingly furamidine was shown globally to rescue many mis-splicing events with only modest off-target effects on gene expression in the HSALR DM1 mouse model. Importantly, over 20% of the differentially expressed genes were shown to be returned, to varying degrees, to wild-type expression levels.


Subject(s)
Benzamidines/therapeutic use , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/genetics , RNA Splicing/drug effects , Animals , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Benzamidines/pharmacology , Cells, Cultured , Disease Models, Animal , Humans , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , RNA/genetics , RNA/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
20.
Int J Biol Macromol ; 119: 1113-1128, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30098361

ABSTRACT

Development of resistance against existing anti-epileptic drugs has alarmed the scientific innovators to find novel potential chemical starting points for the treatment of epilepsy and GABAA inhibition is a promising drug target strategy against epilepsy. The crystal structure of a subtype-selective ß3-homopentameric ligand-gated ion channel of GABAA receptor has been used for the first time for screening the Asinex library for discovery of GABAA agonists as potential anti-epileptic agents. Co-crystallized ligand established the involvement of part of the ß7-ß8 loop (Glu155 and Tyr157) and ß9-ß10 loop (Phe200 and Tyr205) residues as the crucial amino acids in effective binding, an essential feature, being hydrogen bond or ionic interaction with Glu155 residue. Top ranked hits were further subjected to binding energy estimation, ADMET analysis and ligand efficiency matric calculations as consecutive filters. About 19 compounds qualifying all parameters possessed interaction of one positively charged group with Glu155 with good CNS drug-like properties. Simulation studies were performed on the apo protein, its complex with co-crystallized ligand and the best hit qualifying all screening parameters. The best hit was also analyzed using Quantum mechanical studies, off-target analysis and hit modification. The off-target analysis emphasized that these agents did not have any other predicted side-effects.


Subject(s)
Epilepsy/drug therapy , GABA-A Receptor Agonists/chemistry , GABA-A Receptor Agonists/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , Quantum Theory , Receptors, GABA-A/metabolism , Benzamidines/chemistry , Benzamidines/metabolism , Benzamidines/pharmacology , Benzamidines/therapeutic use , Drug Evaluation, Preclinical , GABA-A Receptor Agonists/metabolism , GABA-A Receptor Agonists/therapeutic use , Humans , Ligands , Protein Conformation , Receptors, GABA-A/chemistry , Structure-Activity Relationship , User-Computer Interface
SELECTION OF CITATIONS
SEARCH DETAIL
...