197 results on '"camostat"'
Search Results
2. Favipiravir, camostat, and ciclesonide combination therapy in patients with moderate COVID-19 pneumonia with/without oxygen therapy: An open-label, single-center phase 3 randomized clinical trial
- Author
-
Terada, Jiro, Fujita, Retsu, Kawahara, Takuya, Hirasawa, Yasutaka, Kinoshita, Taku, Takeshita, Yuichiro, Isaka, Yuri, Kinouchi, Toru, Tajima, Hiroshi, Tada, Yuji, and Tsushima, Kenji
- Published
- 2022
- Full Text
- View/download PDF
3. Computational screening of camostat and related compounds against human TMPRSS2: A potential treatment of COVID-19
- Author
-
Sharma, Tanuj, Baig, Mohammad Hassan, Khan, Mohd Imran, Alotaibi, Saqer S., Alorabi, Mohammed, and Dong, Jae-June
- Published
- 2022
- Full Text
- View/download PDF
4. Camostat mesylate inhibits SARS-CoV-2 activation by TMPRSS2-related proteases and its metabolite GBPA exerts antiviral activity
- Author
-
Hoffmann, Markus, Hofmann-Winkler, Heike, Smith, Joan C., Krüger, Nadine, Arora, Prerna, Sørensen, Lambert K., Søgaard, Ole S., Hasselstrøm, Jørgen Bo, Winkler, Michael, Hempel, Tim, Raich, Lluís, Olsson, Simon, Danov, Olga, Jonigk, Danny, Yamazoe, Takashi, Yamatsuta, Katsura, Mizuno, Hirotaka, Ludwig, Stephan, Noé, Frank, Kjolby, Mads, Braun, Armin, Sheltzer, Jason M., and Pöhlmann, Stefan
- Published
- 2021
- Full Text
- View/download PDF
5. Camostat Mesilate Treating Patients With Hospitalized Patients With COVID-19 (RECOVER)
- Author
-
Alan Bryce, Principal Investigator
- Published
- 2024
6. ACTIV-2: A Study for Outpatients With COVID-19
- Author
-
Eli Lilly and Company, Advancing Clinical Therapeutics Globally for HIV/AIDS and Other Infections, Brii Biosciences Limited, AstraZeneca, Sagent Pharmaceuticals, Synairgen Research Ltd., Bristol-Myers Squibb, and SAb Biotherapeutics, Inc.
- Published
- 2024
7. The Potential of Camostat in COVID-19 (COV-AAT)
- Published
- 2024
8. Multiple-dose Study of FOY-305 in Japanese Healthy Adult Male Subjects
- Published
- 2024
9. A Study of FOY-305 in Patients With SARS-Cov-2 Infection (COVID-19)
- Published
- 2024
10. One Week of Oral Camostat Versus Placebo in Nonhospitalized Adults With Mild-to-Moderate Coronavirus Disease 2019: A Randomized Controlled Phase 2 Trial
- Author
-
Jilg, Nikolaus, Chew, Kara W, Giganti, Mark J, Daar, Eric S, Wohl, David A, Javan, Arzhang Cyrus, Kantor, Amy, Moser, Carlee, Coombs, Robert W, Neytman, Gene, Hoover, Keila, Jana, Atasi, Hart, Phil A, Greninger, Alexander L, Szurgot, Bob, Eron, Joseph J, Currier, Judith S, Hughes, Michael D, Smith, Davey M, Li, Jonathan Z, Chew, Kara, Smith, David, Daar, Eric, Wohl, David, Currier, Judith, Eron, Joseph, Hughes, Michael, Giganti, Mark, Ritz, Justin, Hosey, Lara, Roa, Jhoanna, Patel, Nilam, Colsh, Kelly, Rwakazina, Irene, Beck, Justine, Sieg, Scott, Li, Jonathan, Fletcher, Courtney, Fischer, William, Evering, Teresa, Coombs, Robert, Ignacio, Rachel Bender, Cardoso, Sandra, Corado, Katya, Jagannathan, Prasanna, Perelson, Alan, Pillay, Sandy, Riviere, Cynthia, Singh, Upinder, Taiwo, Babafemi, Gottesman, Joan, Newell, Matthew, Pedersen, Susan, Dragavon, Joan, Jennings, Cheryl, Greenfelder, Brian, Murtaugh, William, Kosmyna, Jan, Gapara, Morgan, and Shahkolahi, Akbar
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Infectious Diseases ,Coronaviruses Therapeutics and Interventions ,Coronaviruses ,Emerging Infectious Diseases ,Clinical Trials and Supportive Activities ,Clinical Research ,6.1 Pharmaceuticals ,Good Health and Well Being ,Humans ,Adult ,COVID-19 ,SARS-CoV-2 ,RNA ,Viral ,Time Factors ,Treatment Outcome ,camostat ,outpatient ,phase 2 ,ACTIV-2/A5401 Study Team ,Biological Sciences ,Medical and Health Sciences ,Microbiology ,Clinical sciences - Abstract
BackgroundCamostat inhibits severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in vitro. We studied the safety and efficacy of camostat in ACTIV-2/A5401, a phase 2/3 platform trial of therapeutics for COVID-19 in nonhospitalized adults.MethodsWe conducted a phase 2 study in adults with mild-to-moderate COVID-19 randomized to oral camostat for 7 days or a pooled placebo arm. Primary outcomes were time to improvement in COVID-19 symptoms through day 28, proportion of participants with SARS-CoV-2 RNA below the lower limit of quantification (LLoQ) from nasopharyngeal swabs through day 14, and grade ≥3 treatment-emergent adverse events (TEAEs) through day 28.ResultsOf 216 participants (109 randomized to camostat, 107 to placebo) who initiated study intervention, 45% reported ≤5 days of symptoms at study entry and 26% met the protocol definition of higher risk of progression to severe COVID-19. Median age was 37 years. Median time to symptom improvement was 9 days in both arms (P = .99). There were no significant differences in the proportion of participants with SARS-CoV-2 RNA ConclusionsIn a phase 2 study of nonhospitalized adults with mild-to-moderate COVID-19, oral camostat did not accelerate viral clearance or time to symptom improvement, or reduce hospitalizations or deaths. Clinical Trials Registration. ClinicalTrials.gov identifier: NCT04518410.
- Published
- 2023
11. A Trial Looking at the Use of Camostat in People Who Have Tested Positive for Coronavirus (COVID-19) (SPIKE-1) (SPIKE-1)
- Author
-
Latus Therapeutics
- Published
- 2023
12. Camostat With Bicalutamide for COVID-19 (COMBO)
- Published
- 2023
13. COVID-19 Outpatient Pragmatic Platform Study (COPPS) - Camostat Sub-Protocol (COPPS)
- Published
- 2023
14. COVID-19 Outpatient Pragmatic Platform Study (COPPS) - Master Protocol
- Published
- 2023
15. The DAWN Antivirals Trial for Ambulatory COVID-19 Patients (DAWN)
- Author
-
Universiteit Antwerpen, University Ghent, Université de Liège, Vrije Universiteit Brussel, and Ann Van den Bruel, Professor
- Published
- 2022
16. Camostat Mesylate for Protein-losing Enteropathy After Fontan Operation
- Author
-
Gi Beom Kim, Professor
- Published
- 2022
17. Oral Camostat Compared With Standard Supportive Care in Mild-Moderate COVID-19 Patients (COPS-2003)
- Published
- 2022
18. The Utility of Camostat Mesylate in Patients With COVID-19 Associated Coagulopathy (CAC) and Cardiovascular Complications
- Author
-
Ono Pharmaceutical Co. Ltd and Arya Mani, Professor of Medicine and of Genetics
- Published
- 2022
19. Camostat Mesylate in COVID-19 Outpatients
- Published
- 2022
20. Reconvalescent Plasma/Camostat Mesylate Early in SARS-CoV-2 Q-PCR (COVID-19) Positive High-risk Individuals (RES-Q-HR)
- Author
-
The Federal Ministry of Health, Germany (Bundesministerium für Gesundheit, BMG)
- Published
- 2022
21. CAMOVID : Evaluation of Efficacy and Safety of Camostat Mesylate for the Treatment of SARS-CoV-2 Infection - COVID-19 in Ambulatory Adult Patients (CAMOVID)
- Published
- 2022
22. Novel Agents for Treatment of High-risk COVID-19 Positive Patients
- Author
-
Susanne Arnold, Professor
- Published
- 2022
23. Camostat Efficacy vs. Placebo for Outpatient Treatment of COVID-19 (CAMELOT)
- Published
- 2022
24. Safety and Preliminary Efficacy of the Combination of Niclosamide and Camostat (NICCAM)
- Author
-
Bayer
- Published
- 2021
25. Camostat and Artemisia Annua vs Placebo in COVID-19 Outpatients
- Published
- 2021
26. The Impact of Camostat Mesilate on COVID-19 Infection (CamoCO-19)
- Published
- 2021
27. Efficacy and safety of camostat mesylate in early COVID-19 disease in an ambulatory setting: a randomized placebo-controlled phase II trial
- Author
-
Els Tobback, Sophie Degroote, Sabine Buysse, Liesbeth Delesie, Lucas Van Dooren, Sophie Vanherrewege, Cyril Barbezange, Veronik Hutse, Marta Romano, Isabelle Thomas, Elizaveta Padalko, Steven Callens, and Marie-Angélique De Scheerder
- Subjects
Camostat ,COVID-19 ,Efficacy ,Neutralizing antibodies ,Randomized controlled trial ,Safety ,Infectious and parasitic diseases ,RC109-216 - Abstract
Objectives: This study aimed to assess the efficacy and safety of 300 mg camostat mesylate three times daily in a fasted state to treat early phase COVID-19 in an ambulatory setting. Methods: We conducted a phase II randomized controlled trial in symptomatic (maximum 5 days) and asymptomatic patients with confirmed COVID-19 infection. Patients were randomly assigned in a 2:1 ratio to receive either camostat mesylate or a placebo. Outcomes included change in nasopharyngeal viral load, time to clinical improvement, the presence of neutralizing antibodies, and safety. Results: Of 96 participants randomized between November 2020 and June 2021, analyses were performed on the data of 90 participants who completed treatment (N = 61 camostat mesylate, N = 29 placebo). The estimated mean change in cycle threshold between day 1 and day 5 between the camostat and placebo group was 1.183 (P = 0.511). The unadjusted hazard ratio for clinical improvement in the camostat group was 0.965 (95% confidence interval, 0.480-1.942, P = 0.921 by Cox regression). The percentage distribution of the 50% neutralizing antibody titer at day 28 visit and frequency of adverse events were similar between the two groups. Conclusion: Under this protocol, camostat mesylate was not found to be effective as an antiviral drug against SARS-CoV-2.Trial registration: ClinicalTrials.gov NCT04625114; November 12, 2020.
- Published
- 2022
- Full Text
- View/download PDF
28. Combination Therapy With Camostat Mesilate + Hydroxychloroquine for COVID-19 (CLOCC)
- Author
-
Universitätsklinikum Hamburg-Eppendorf, Goethe University, St. Georg Hospital Leipzig, Germany, Hospital Schwabing Munich, Germany, and Missioklinik, Wuerzburg, Germany
- Published
- 2020
29. Open Label Study to Compare Efficacy, Safety and Tolerability of Hydroxychloroquine Combined With Azithromycin Compared to Hydroxychloroquine Combined With Camostat Mesylate and to 'no Treatment' in SARS CoV 2 Virus (COSTA)
- Author
-
Itsik Levy Dr, Infectious Disease specialist
- Published
- 2020
30. 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
31. Computational screening of camostat and related compounds against human TMPRSS2: A potential treatment of COVID-19
- Author
-
Tanuj Sharma, Mohammad Hassan Baig, Mohd Imran Khan, Saqer S. Alotaibi, Mohammed Alorabi, and Jae-June Dong
- Subjects
Severe acute respiratory syndrome coronavirus 2 ,Main protease ,Camostat ,Inhibitors ,Therapeutics. Pharmacology ,RM1-950 - Abstract
The global coronavirus pandemic has burdened the human population with mass fatalities and disastrous socio-economic consequences. The frequent occurrence of these new variants has fueled the already prevailing challenge. There is still a necessity for highly effective small molecular agents to prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Here, we targeted the human transmembrane surface protease TMPRSS2, which is essential for proteolytic activation of SARS-CoV-2. Camostat is a well-known inhibitor of serine proteases and an effective TMPRSS2 inhibitor. A virtual library of camostat-like compounds was computationally screened against the catalytic site of TMPRSS2. Following a sequential in-depth molecular docking and dynamics simulation, we report the compounds that exhibited promising efficacy against TMPRSS2. The molecular docking and MM/PBSA free energy calculation study indicates these compounds carry excellent binding affinity against TMPRSS2 and found them more effective than camostat. The study will open doors for the effective treatment of coronavirus disease 2019.
- Published
- 2022
- Full Text
- View/download PDF
32. Camostat Does Not Inhibit the Proteolytic Activity of Neutrophil Serine Proteases.
- Author
-
Assylbekova, Akmaral, Zhanapiya, Anuar, Grzywa, Renata, Sienczyk, Marcin, Schönbach, Christian, and Burster, Timo
- Subjects
- *
ELASTASES , *SARS-CoV-2 , *TRYPSIN , *SERINE proteinases , *CORONAVIRUS spike protein , *COVID-19 , *LEUCOCYTE elastase - Abstract
Coronavirus disease 2019 (COVID-19) can lead to multi-organ failure influenced by comorbidities and age. Binding of the severe acute respiratory syndrome coronavirus 2 spike protein (SARS-CoV-2 S protein) to angiotensin-converting enzyme 2 (ACE2), along with proteolytic digestion of the S protein by furin and transmembrane protease serine subtype 2 (TMPRSS2), provokes internalization of SARS-CoV-2 into the host cell. Productive infection occurs through viral replication in the cytosol and cell-to-cell transmission. The catalytic activity of TMPRSS2 can be blocked by the trypsin-like serine protease inhibitor camostat, which impairs infection by SARS-CoV-2. At the site of infection, immune cells, such as neutrophils, infiltrate and become activated, releasing neutrophil serine proteases (NSPs), including cathepsin G (CatG), neutrophil elastase (NE), and proteinase 3 (PR3), which promote the mounting of a robust immune response. However, NSPs might be involved in infection and the severe outcome of COVID-19 since the uncontrolled proteolytic activity is responsible for many complications, including autoimmunity, chronic inflammatory disorders, cardiovascular diseases, and thrombosis. Here, we demonstrate that camostat does not inhibit the catalytic activity of CatG, NE, and PR3, indicating the need for additional selective serine protease inhibitors to reduce the risk of developing severe COVID-19. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
33. Favipiravir, camostat, and ciclesonide combination therapy in patients with moderate COVID-19 pneumonia with/without oxygen therapy: An open-label, single-center phase 3 randomized clinical trial
- Author
-
Jiro Terada, Retsu Fujita, Takuya Kawahara, Yasutaka Hirasawa, Taku Kinoshita, Yuichiro Takeshita, Yuri Isaka, Toru Kinouchi, Hiroshi Tajima, Yuji Tada, and Kenji Tsushima
- Subjects
COVID-19 ,Camostat ,Ciclesonide ,Combination therapy ,Favipiravir ,Hospital stay ,Medicine (General) ,R5-920 - Abstract
Summary: Background: The effectiveness of combination therapy for COVID-19 pneumonia remains unclear. We evaluated favipiravir, camostat, and ciclesonide combination therapy in patients with moderate COVID-19 pneumonia. Methods: In this open-label phase 3 study, hospitalized adults who were positive for SARS-CoV-2 and had COVID-19 pneumonia were enrolled prior to official vaccination drive in Japan. Participants were randomly assigned to favipiravir monotherapy or favipiravir + camostat + ciclesonide combination therapy. The primary outcome was the length of hospitalization due to COVID-19 infection after study treatment. The hospitalization period was calculated from the time of admission to the time of patient discharge using the clinical management guide of COVID-19 for front-line healthcare workers developed by the Japanese Ministry of Health, Labour, and Welfare (Version 3). Cases were registered between November 11, 2020, and May 31, 2021. Japan Registry of Clinical Trials registration: jRCTs031200196. Findings: Of 121 enrolled patients, 56 received monotherapy and 61 received combination therapy. Baseline characteristics were balanced between the groups. The median time of hospitalization was 10 days for the combination and 11 days for the monotherapy group. The median time to discharge was statistically significantly lower in the combination therapy vs monotherapy group (HR, 1·67 (95% CI 1·03–2·7; P = 0·035). The hospital discharge rate was statistically significantly higher in the combination therapy vs monotherapy group in patients with less severe COVID-19 infections and those who were ≤60 years. There were no significant differences in clinical findings between the groups at 4, 8, 11, 15, and 29 days. Adverse events were comparable between the groups. There were two deaths, with one in each group. Interpretation: Combination oral favipiravir, camostat and, ciclesonide therapy could decrease the length of hospitalization stays without safety concerns in patients with moderate COVID-19 pneumonia. However, lack of hard clinical primary outcome is one of the major limitations of the study. Funding: This research was supported by Japan Agency for Medical Research and Development (AMED) under Grant Number 20fk0108261h0001.
- Published
- 2022
- Full Text
- View/download PDF
34. SPIKE-1: A Randomised Phase II/III trial in a community setting, assessing use of camostat in reducing the clinical progression of COVID-19 by blocking SARS-CoV-2 Spike protein-initiated membrane fusion
- Author
-
Sarah Halford, Susan Wan, Ilaria Dragoni, Julie Silvester, Bobojon Nazarov, Daniel Anthony, Suzie Anthony, Emma Ladds, John Norrie, Kevin Dhaliwal, and the CDD SPIKE-1 Project Team
- Subjects
COVID-19 ,Randomised controlled trial ,protocol ,camostat ,TMPRSS2 ,Spike ,Medicine (General) ,R5-920 - Abstract
Abstract Objectives The primary objective is to evaluate the efficacy of camostat to prevent respiratory deterioration in patients with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection. Secondary objectives include assessment of the ability of camostat to reduce the requirement for Coronavirus disease 2019 (COVID-19) related hospital admission and to reduce the requirement for supplementary oxygen and ventilation as treatment for SARS-CoV-2 infection, to evaluate overall mortality related to COVID-19 and to evaluate the efficacy of camostat by effect on clinical improvement. Research objectives include to assess change in COVID-19 symptom severity, to evaluate the ability of camostat to reduce viral load throughout duration of illness as well as translational research on host and viral genomics, serum antibody production, COVID-19 diagnostics, and validation of laboratory testing methods and biomarkers. Trial design SPIKE-1 is a randomised, multicentre, prospective, open label, community-based clinical trial. Eligible patients will be randomised 1:1 to the camostat treatment arm and control arm (best supportive care). The trial is designed to include a pilot phase recruiting up to 50 patients in each arm. An initial review at the end of the pilot phase will allow assessment of available data and inform the requirement for any protocol adaptations to include refinement of eligibility criteria to enrich the patient population and sample size calculations. Up to 289 additional patients will be randomised in the continuation phase of the trial. A formal interim analysis will be performed once 50% of the maximum sample size has been recruited Participants The trial will recruit adults (≥ 18 years) who score moderate to very high risk according to COVID-age risk calculation, with typical symptoms of COVID-19 infection as per Public Health England guidance or equivalent organisations in the UK, Health Protection Scotland, Public Health Wales, Public Health Agency (Northern Ireland) and with evidence of current COVID-19 infection from a validated assay. The trial is being conducted in the UK and patients are recruited through primary care and hospital settings. Intervention and comparator Eligible patients with be randomised to receive either camostat tablets, 200 mg four times daily (qds) for 14 days (treatment arm) or best supportive care (control arm). Main outcomes Primary outcome measure: the rate of hospital admissions requiring supplemental oxygen. Secondary outcome measures include: the rate of COVID-19 related hospital admission in patients with SARS-CoV-2 infection; the number of supplementary oxygen-free days and ventilator-free days measured at 28 days from randomisation; the rate of mortality related to COVID-19 one year from randomisation; the time to worst point on the nine-point category ordinal scale (recommended by the World Health Organization: Coronavirus disease (COVID-2019)) or deterioration of two points or more, within 28 days from randomisation. Research outcomes include the assessment of change in COVID-19 symptom severity on days 1-14 as measured by (1) time to apyrexia (maintained for 48 hrs) by daily self-assessment of temperature, time to improvement (by two points) in peripheral oxygenation saturation defined by daily self-assessment of fingertip peripheral oxygenation saturation levels, (3) assessment of COVID-19 symptoms using the Flu-iiQ questionnaire (determined by app recording and/or daily video call (or phone) consultation and (4) assessment of functional score (where possible) at screening, day 7 and 14. The ability of camostat to reduce viral load throughout duration of illness will be assessed by (1) change in respiratory (oropharyngeal/nasopharyngeal swab RT-PCR) log10 viral load from baseline to Days 7 and 14, (2) change in respiratory (saliva RT-PCR) log10 viral load from baseline to Days 1-14 and (3) change in upper respiratory viral shedding at Day 1 -14 measured as time to clearance of nasal SARS-CoV-2, defined as 2 consecutive negative swabs by qPCR. Additional translational research outcomes include assessment of host and viral genomics, serum antibody production and COVID-19 diagnostics at baseline and on Days 7 and 14. Randomisation Eligible patients will be randomised using an interactive web response system (IWRS) in a 1:1 ratio to one of two arms: (1) treatment arm or (2) control arm. Blinding (masking) The trial is open-label. Numbers to be randomised (sample size) The trial is designed to include a pilot and a continuation phase. Up to 100 patients (randomised 1:1 treatment and control arm) will be recruited in the pilot phase and a maximum of 289 patients (randomised 1:1 treatment and control) will be recruited as part of the continuation phase. The total number of patients recruited will not exceed 389. Trial Status Protocol version number v3 25 September 2020. Trial opened to recruitment on 04 August 2020. The authors anticipate recruitment to be completed by October 2021. Trial registration EudraCT 2020-002110-41; 18 June 2020 ClinicalTrials.gov NCT04455815 ; 02 July 2020 Full protocol The full protocol is attached as an additional file, accessible from the Trials website (Additional file 1). Unpublished PK data provided under confidentiality agreement to the trial Sponsor has been removed from the background section of the protocol to allow for publication of the trial protocol. In the interest in expediting dissemination of this material, the familiar formatting has been eliminated; this Letter serves as a summary of the key elements of the full protocol.
- Published
- 2021
- Full Text
- View/download PDF
35. Semi-Mechanistic Pharmacokinetic-Pharmacodynamic Model of Camostat Mesylate-Predicted Efficacy against SARS-CoV-2 in COVID-19
- Author
-
Yuri Kosinsky, Kirill Peskov, Donald R. Stanski, Diana Wetmore, and Joseph Vinetz
- Subjects
COVID-19 ,antiviral pharmacology ,camostat ,Microbiology ,QR1-502 - Abstract
ABSTRACT The SARS-CoV-2 coronavirus, which causes COVID-19, uses a viral surface spike protein for host cell entry and the human cell-surface transmembrane serine protease, TMPRSS2, to process the spike protein. Camostat mesylate, an orally available and clinically used serine protease inhibitor, inhibits TMPRSS2, supporting clinical trials to investigate its use in COVID-19. A one-compartment pharmacokinetic (PK)/pharmacodynamic (PD) model for camostat and the active metabolite FOY-251 was developed, incorporating TMPRSS2 reversible covalent inhibition by FOY-251, and empirical equations linking TMPRSS2 inhibition of SARS-CoV-2 cell entry. The model predicts that 95% inhibition of TMPRSS2 is required for 50% inhibition of viral entry efficiency. For camostat 200 mg dosed four times daily, 90% inhibition of TMPRSS2 is predicted to occur but with only about 40% viral entry inhibition. For 3-fold higher camostat dosing, marginal improvement of viral entry rate inhibition, up to 54%, is predicted. Because respiratory tract viral load may be associated with negative outcome, even modestly reducing viral entry and respiratory tract viral load may reduce disease progression. This modeling also supports medicinal chemistry approaches to enhancing PK/PD and potency of the camostat molecule. IMPORTANCE Strategies to repurpose already-approved drugs for the treatment of COVID-19 has been attractive since the beginning of the pandemic. Camostat mesylate, a serine protease inhibitor approved in Japan for the treatment of acute exacerbations of chronic pancreatitis, inhibits TMPRSS1, a host cell surface serine protease essential for SARS-CoV-2 viral entry. In vitro experiments provided data suggesting that camostat might be effective in the treatment of COVID-19. Multiple clinical trials were planned to test the hypothesis that camostat would be beneficial for treating COVID-19 (for example, clinicaltrials.gov, NCT04353284). The present work used a one-compartment pharmacokinetic (PK)/pharmacodynamic (PD) mathematical model for camostat and the active metabolite FOY-251, incorporating TMPRSS2 reversible covalent inhibition by FOY-251, and empirical equations linking TMPRSS2 inhibition of SARS-CoV-2 cell entry. This work is valuable to guide further development of camostat mesylate and possible medicinal chemistry derivatives for the treatment of COVID-19.
- Published
- 2022
- Full Text
- View/download PDF
36. The TMPRSS2 Inhibitor Nafamostat Reduces SARS-CoV-2 Pulmonary Infection in Mouse Models of COVID-19
- Author
-
Kun Li, David K. Meyerholz, Jennifer A. Bartlett, and Paul B. McCray
- Subjects
COVID-19 ,SARS-CoV-2 ,MERS-CoV ,nafamostat ,camostat ,TMPRSS2 ,Microbiology ,QR1-502 - Abstract
ABSTRACT The coronavirus disease 2019 (COVID-19) pandemic has caused significant morbidity and mortality on a global scale. The etiologic agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), initiates host cell entry when its spike protein (S) binds to its receptor, angiotensin-converting enzyme 2 (ACE2). In airway epithelia, the spike protein is cleaved by the cell surface protease TMPRSS2, facilitating membrane fusion and entry at the cell surface. This dependence on TMPRSS2 and related proteases suggests that protease inhibitors might limit SARS-CoV-2 infection in the respiratory tract. Here, we tested two serine protease inhibitors, camostat mesylate and nafamostat mesylate, for their ability to inhibit entry of SARS-CoV-2 and that of a second pathogenic coronavirus, Middle East respiratory syndrome coronavirus (MERS-CoV). Both camostat and nafamostat reduced infection in primary human airway epithelia and in the Calu-3 2B4 cell line, with nafamostat exhibiting greater potency. We then assessed whether nafamostat was protective against SARS-CoV-2 in vivo using two mouse models. In mice sensitized to SARS-CoV-2 infection by transduction with human ACE2, intranasal nafamostat treatment prior to or shortly after SARS-CoV-2 infection significantly reduced weight loss and lung tissue titers. Similarly, prophylactic intranasal treatment with nafamostat reduced weight loss, viral burden, and mortality in K18-hACE2 transgenic mice. These findings establish nafamostat as a candidate for the prevention or treatment of SARS-CoV-2 infection and disease pathogenesis. IMPORTANCE The causative agent of COVID-19, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), requires host cell surface proteases for membrane fusion and entry into airway epithelia. We tested the hypothesis that inhibitors of these proteases, the serine protease inhibitors camostat and nafamostat, block infection by SARS-CoV-2. We found that both camostat and nafamostat reduce infection in human airway epithelia, with nafamostat showing greater potency. We then asked whether nafamostat protects mice against SARS-CoV-2 infection and subsequent COVID-19 lung disease. We performed infections in mice made susceptible to SARS-CoV-2 infection by introducing the human version of ACE2, the SARS-CoV-2 receptor, into their airway epithelia. We observed that pretreating these mice with nafamostat prior to SARS-CoV-2 infection resulted in better outcomes, in the form of less virus-induced weight loss, viral replication, and mortality than that observed in the untreated control mice. These results provide preclinical evidence for the efficacy of nafamostat in treating and/or preventing COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
37. Structural Basis of Covalent Inhibitory Mechanism of TMPRSS2- Related Serine Proteases by Camostat.
- Author
-
Gaohui Sun, Yaqun Sui, Yang Zhou, Junlin Ya, Cai Yuan, Longguang Jiang, and Mingdong Huang
- Subjects
- *
SERINE proteinases , *COVID-19 , *COVID-19 treatment , *VIRAL transmission , *PLASMINOGEN activators , *PLASMINOGEN , *PROTEOLYTIC enzymes - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the viral pathogen causing the coronavirus disease 2019 (COVID-19) global pandemic. No effective treatment for COVID-19 has been established yet. The serine protease transmembrane protease serine 2 (TMPRSS2) is essential for viral spread and pathogenicity by facilitating the entry of SARS-CoV-2 into host cells. The protease inhibitor camostat, an anticoagulant used in the clinic, has potential anti-inflammatory and antiviral activities against COVID-19. However, the potential mechanisms of viral resistance and antiviral activity of camostat are unclear. Herein, we demonstrate high inhibitory potencies of camostat for a panel of serine proteases, indicating that camostat is a broad-spectrum inhibitor of serine proteases. In addition, we determined the crystal structure of camostat in complex with a serine protease (uPA [urokinase-type plasminogen activator]), which reveals that camostat is inserted in the S1 pocket of uPA but is hydrolyzed by uPA, and the cleaved camostat covalently binds to Ser195. We also generated a homology model of the structure of the TMPRSS2 serine protease domain. The model shows that camostat uses the same inhibitory mechanism to inhibit the activity of TMPRSS2, subsequently preventing SARS-CoV-2 spread [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
38. A phase 1/2 trial to evaluate the pharmacokinetics, safety, and efficacy of NI-03 in patients with chronic pancreatitis: study protocol for a randomized controlled trial on the assessment of camostat treatment in chronic pancreatitis (TACTIC)
- Author
-
Mitchell L. Ramsey, Janet Nuttall, Phil A. Hart, and on behalf of the TACTIC Investigative Team
- Subjects
Pancreatic diseases ,Abdominal pain ,Serine protease inhibitor ,Camostat ,Medicine (General) ,R5-920 - Abstract
Abstract Background Chronic pancreatitis (CP) is a progressive, fibro-inflammatory disease characterized by enzymatic autoactivation and subsequent fibrotic replacement of acinar cells. A significant proportion of patients develop pain, which may be due to many causes, including perineural inflammation, altered central processing of pain signals, parenchymal structural changes, and ductal obstruction. Currently there are no approved medical treatment options for CP-associated pain. NI-03 (camostat mesilate) is an orally administered serine protease inhibitor that reduces pancreatic enzyme activity and has been widely used for the treatment of CP-associated pain in Japan. The current study will assess the safety and efficacy of NI-03 for reduction of CP-associated pain in the USA. Methods The current study consists of two phases. First, a phase I study will be performed to establish the pharmacokinetics and safety profile over a 1-week period following a single dose (100, 200, or 300 mg). Subsequently, a phase II study will be performed consisting of a double-blind, randomized, controlled trial (RCT). This RCT will evaluate the efficacy of each of the three doses of NI-03 given three times daily compared to placebo over 28 days. A 7-day, single-blind, run-in period will precede the double-blind phase to assess baseline pain characteristics. The primary efficacy outcome is the average of worst daily pain scores (numeric rating scale of 0–10) over the terminal 7 days of the study period compared to baseline. Secondary efficacy outcomes include change in opioid dose and quality of life measures, and time to first rescue intravenous analgesic. Adverse events will be recorded. Discussion NI-03 has been used successfully and safely in Japan to treat CP-associated pain. The aim of the current study is to assess the safety and efficacy of NI-03 using a rigorous RCT in a population in the USA. This study may fill an important clinical gap to provide an effective medical treatment option for CP-associated pain. Trial registration ClinicalTrials.gov, NCT02693093. Registered through the National Institutes of Health on 26 February 2016.
- Published
- 2019
- Full Text
- View/download PDF
39. Camostat Does Not Inhibit the Proteolytic Activity of Neutrophil Serine Proteases
- Author
-
Akmaral Assylbekova, Anuar Zhanapiya, Renata Grzywa, Marcin Sienczyk, Christian Schönbach, and Timo Burster
- Subjects
serine proteases ,cathepsin G ,neutrophil elastase ,proteinase 3 ,camostat ,SARS-CoV-2 ,Medicine ,Pharmacy and materia medica ,RS1-441 - Abstract
Coronavirus disease 2019 (COVID-19) can lead to multi-organ failure influenced by comorbidities and age. Binding of the severe acute respiratory syndrome coronavirus 2 spike protein (SARS-CoV-2 S protein) to angiotensin-converting enzyme 2 (ACE2), along with proteolytic digestion of the S protein by furin and transmembrane protease serine subtype 2 (TMPRSS2), provokes internalization of SARS-CoV-2 into the host cell. Productive infection occurs through viral replication in the cytosol and cell-to-cell transmission. The catalytic activity of TMPRSS2 can be blocked by the trypsin-like serine protease inhibitor camostat, which impairs infection by SARS-CoV-2. At the site of infection, immune cells, such as neutrophils, infiltrate and become activated, releasing neutrophil serine proteases (NSPs), including cathepsin G (CatG), neutrophil elastase (NE), and proteinase 3 (PR3), which promote the mounting of a robust immune response. However, NSPs might be involved in infection and the severe outcome of COVID-19 since the uncontrolled proteolytic activity is responsible for many complications, including autoimmunity, chronic inflammatory disorders, cardiovascular diseases, and thrombosis. Here, we demonstrate that camostat does not inhibit the catalytic activity of CatG, NE, and PR3, indicating the need for additional selective serine protease inhibitors to reduce the risk of developing severe COVID-19.
- Published
- 2022
- Full Text
- View/download PDF
40. In Silico Analysis and Synthesis of Nafamostat Derivatives and Evaluation of Their Anti-SARS-CoV-2 Activity
- Author
-
Kazuhiro J. Fujimoto, Daniel C. F. Hobbs, Miki Umeda, Akihiro Nagata, Rie Yamaguchi, Yoshitaka Sato, Ayato Sato, Kohsuke Ohmatsu, Takashi Ooi, Takeshi Yanai, Hiroshi Kimura, and Takayuki Murata
- Subjects
COVID-19 ,anti-SARS-CoV-2 agent ,TMPRSS2 ,nafamostat ,camostat ,Microbiology ,QR1-502 - Abstract
Inhibition of transmembrane serine protease 2 (TMPRSS2) is expected to block the spike protein-mediated fusion of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Nafamostat, a potent TMPRSS2 inhibitor as well as a candidate for anti-SARS-CoV-2 drug, possesses the same acyl substructure as camostat, but is known to have a greater antiviral effect. A unique aspect of the molecular binding of nafamostat has been recently reported to be the formation of a covalent bond between its acyl substructure and Ser441 in TMPRSS2. In this study, we investigated crucial elements that cause the difference in anti-SARS-CoV-2 activity of nafamostat and camostat. In silico analysis showed that Asp435 significantly contributes to the binding of nafamostat and camostat to TMPRSS2, while Glu299 interacts strongly only with nafamostat. The estimated binding affinity for each compound with TMPRSS2 was actually consistent with the higher activity of nafamostat; however, the evaluation of the newly synthesized nafamostat derivatives revealed that the predicted binding affinity did not correlate with their anti-SARS-CoV-2 activity measured by the cytopathic effect (CPE) inhibition assay. It was further shown that the substitution of the ester bond with amide bond in nafamostat resulted in significantly weakened anti-SARS-CoV-2 activity. These results strongly indicate that the ease of covalent bond formation with Ser441 in TMPRSS2 possibly plays a major role in the anti-SARS-CoV-2 effect of nafamostat and its derivatives.
- Published
- 2022
- Full Text
- View/download PDF
41. Protease Inhibitors: Candidate Drugs to Inhibit Severe Acute Respiratory Syndrome Coronavirus 2 Replication.
- Author
-
Mutsuo Yamaya, Hidekazu Nishimura, Xue Deng, Akiko Kikuchi, and Ryoichi Nagatomi
- Abstract
The number of patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly increased, although the WHO declared a pandemic. However, drugs that function against SARS-CoV-2 have not been established. SARS-CoV-2 has been suggested to bind angiotensin-converting enzyme 2, the receptor of the SARS coronavirus. SARS coronavirus and coronavirus 229E, the cause of the common cold, replicate through cell-surface and endosomal pathways using a protease, the type II transmembrane protease. To examine the effects of protease inhibitors on the replication of coronavirus 229E, we pretreated primary cultures of human nasal epithelial (HNE) cells with camostat or nafamostat, each of which has been used for the treatment of pancreatitis and/or disseminated intravascular coagulation. HNE cells were then infected with coronavirus 229E, and viral titers in the airway surface liquid of the cells were examined. Pretreatment with camostat (0.1-10 µ g/mL) or nafamostat (0.01-1 µ g/mL) reduced the titers of coronavirus 229E. Furthermore, a significant amount of type II transmembrane protease protein was detected in the airway surface liquid of HNE cells. Additionally, interferons have been reported to have antiviral effects against SARS coronavirus. The additive effects of interferons on the inhibitory effects of other candidate drugs to treat SARS-CoV-2 infection, such as lopinavir, ritonavir and favipiravir, have also been studied. These findings suggest that protease inhibitors of this type may inhibit coronavirus 229E replication in human airway epithelial cells at clinical concentrations. Protease inhibitors, interferons or the combination of these drugs may become candidate drugs to inhibit the replication of SARS-CoV-2. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
42. SPIKE-1: A Randomised Phase II/III trial in a community setting, assessing use of camostat in reducing the clinical progression of COVID-19 by blocking SARS-CoV-2 Spike protein-initiated membrane fusion.
- Author
-
Halford, Sarah, Wan, Susan, Dragoni, Ilaria, Silvester, Julie, Nazarov, Bobojon, Anthony, Daniel, Anthony, Suzie, Ladds, Emma, Norrie, John, Dhaliwal, Kevin, and CDD SPIKE-1 Project Team
- Subjects
- *
COVID-19 , *MEMBRANE fusion , *SARS-CoV-2 , *DISEASE progression , *COMMUNITY-based clinical trials , *VIRAL shedding - Abstract
Objectives: The primary objective is to evaluate the efficacy of camostat to prevent respiratory deterioration in patients with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection. Secondary objectives include assessment of the ability of camostat to reduce the requirement for Coronavirus disease 2019 (COVID-19) related hospital admission and to reduce the requirement for supplementary oxygen and ventilation as treatment for SARS-CoV-2 infection, to evaluate overall mortality related to COVID-19 and to evaluate the efficacy of camostat by effect on clinical improvement. Research objectives include to assess change in COVID-19 symptom severity, to evaluate the ability of camostat to reduce viral load throughout duration of illness as well as translational research on host and viral genomics, serum antibody production, COVID-19 diagnostics, and validation of laboratory testing methods and biomarkers.Trial Design: SPIKE-1 is a randomised, multicentre, prospective, open label, community-based clinical trial. Eligible patients will be randomised 1:1 to the camostat treatment arm and control arm (best supportive care). The trial is designed to include a pilot phase recruiting up to 50 patients in each arm. An initial review at the end of the pilot phase will allow assessment of available data and inform the requirement for any protocol adaptations to include refinement of eligibility criteria to enrich the patient population and sample size calculations. Up to 289 additional patients will be randomised in the continuation phase of the trial. A formal interim analysis will be performed once 50% of the maximum sample size has been recruited PARTICIPANTS: The trial will recruit adults (≥ 18 years) who score moderate to very high risk according to COVID-age risk calculation, with typical symptoms of COVID-19 infection as per Public Health England guidance or equivalent organisations in the UK, Health Protection Scotland, Public Health Wales, Public Health Agency (Northern Ireland) and with evidence of current COVID-19 infection from a validated assay. The trial is being conducted in the UK and patients are recruited through primary care and hospital settings.Intervention and Comparator: Eligible patients with be randomised to receive either camostat tablets, 200 mg four times daily (qds) for 14 days (treatment arm) or best supportive care (control arm).Main Outcomes: Primary outcome measure: the rate of hospital admissions requiring supplemental oxygen. Secondary outcome measures include: the rate of COVID-19 related hospital admission in patients with SARS-CoV-2 infection; the number of supplementary oxygen-free days and ventilator-free days measured at 28 days from randomisation; the rate of mortality related to COVID-19 one year from randomisation; the time to worst point on the nine-point category ordinal scale (recommended by the World Health Organization: Coronavirus disease (COVID-2019)) or deterioration of two points or more, within 28 days from randomisation. Research outcomes include the assessment of change in COVID-19 symptom severity on days 1-14 as measured by (1) time to apyrexia (maintained for 48 hrs) by daily self-assessment of temperature, time to improvement (by two points) in peripheral oxygenation saturation defined by daily self-assessment of fingertip peripheral oxygenation saturation levels, (3) assessment of COVID-19 symptoms using the Flu-iiQ questionnaire (determined by app recording and/or daily video call (or phone) consultation and (4) assessment of functional score (where possible) at screening, day 7 and 14. The ability of camostat to reduce viral load throughout duration of illness will be assessed by (1) change in respiratory (oropharyngeal/nasopharyngeal swab RT-PCR) log10 viral load from baseline to Days 7 and 14, (2) change in respiratory (saliva RT-PCR) log10 viral load from baseline to Days 1-14 and (3) change in upper respiratory viral shedding at Day 1 -14 measured as time to clearance of nasal SARS-CoV-2, defined as 2 consecutive negative swabs by qPCR. Additional translational research outcomes include assessment of host and viral genomics, serum antibody production and COVID-19 diagnostics at baseline and on Days 7 and 14.Randomisation: Eligible patients will be randomised using an interactive web response system (IWRS) in a 1:1 ratio to one of two arms: (1) treatment arm or (2) control arm.Blinding (masking): The trial is open-label.Numbers To Be Randomised (sample Size): The trial is designed to include a pilot and a continuation phase. Up to 100 patients (randomised 1:1 treatment and control arm) will be recruited in the pilot phase and a maximum of 289 patients (randomised 1:1 treatment and control) will be recruited as part of the continuation phase. The total number of patients recruited will not exceed 389.Trial Status: Protocol version number v3 25 September 2020. Trial opened to recruitment on 04 August 2020. The authors anticipate recruitment to be completed by October 2021.Trial Registration: EudraCT 2020-002110-41; 18 June 2020 ClinicalTrials.gov NCT04455815 ; 02 July 2020 FULL PROTOCOL: The full protocol is attached as an additional file, accessible from the Trials website (Additional file 1). Unpublished PK data provided under confidentiality agreement to the trial Sponsor has been removed from the background section of the protocol to allow for publication of the trial protocol. In the interest in expediting dissemination of this material, the familiar formatting has been eliminated; this Letter serves as a summary of the key elements of the full protocol. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
43. Intestinal serine protease inhibition increases FGF21 and improves metabolism in obese mice.
- Author
-
Albarazanji, Kamal, Jennis, Matthew, Cavanaugh, Cassandre R., Lang, Wensheng, Singh, Bhanu, Lanter, James C., Lenhard, James M., and Hornby, Pamela J.
- Abstract
Trypsin is the major serine protease responsible for intestinal protein digestion. An inhibitor, camostat (CS), reduced weight gain, hyperglycemia, and dyslipidemia in obese rats; however, the mechanisms for these are largely unknown. We reasoned that CS creates an apparent dietary protein restriction, which is known to increase hepatic fibroblast growth factor 21 (FGF21). Therefore, metabolic responses to CS and a gut-restricted CS metabolite, FOY-251, were measured in mice. Food intake, body weight, blood glucose, branched-chain amino acids (LC/MS), hormone levels (ELISA), liver pathology (histology), and transcriptional changes (qRT-PCR) were measured in ob/ob, lean and diet-induced obese (DIO) C57BL/6 mice. In ob/ob mice, CS in chow (9-69 mg/kg) or FOY-251 (46 mg/kg) reduced food intake and body weight gain to a similar extent as pair-fed mice. CS decreased blood glucose, liver weight, and lipidosis and increased FGF21 gene transcription and plasma levels. In lean mice, CS increased liver FGF21 mRNA and plasma levels. Relative to pair feeding, FOY-251 also increased plasma FGF21 and induced liver FGF21 and integrated stress response (ISR) transcription. In DIO mice, FOY-251 (100 mg/kg po) did not alter peak glucose levels but reduced the AUC of the glucose excursion in response to an oral glucose challenge. FOY-251 increased plasma FGF21 levels. In addition to previously reported satiety-dependent (cholecystokinin-mediated) actions, intestinal trypsin inhibition engages non-satiety-related pathways in both leptin-deficient and DIO mice. This novel mechanism improves metabolism by a liver-integrated stress response and increased FGF21 expression levels in mice. NEW & NOTEWORTHY Trypsin inhibitors, including plant-based consumer products, have long been associated with metabolic improvements. Studies in the 1980s and 1990s suggested this was due to satiety hormones and caloric wasting by loss of protein and fatty acids in feces. This work suggests an entirely new mechanism based on the lower amounts of digested protein available in the gut. This apparent protein reduction may cause beneficial metabolic adaptation by the intestinal-liver axis to perceived nutrient stress. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
44. Dual inhibition of TMPRSS2 and Cathepsin Bprevents SARS-CoV-2 infection in iPS cells
- Author
-
Emi Sano, Rina Hashimoto, Ayaka Sakamoto, Akitsu Hotta, Shinya Yamanaka, Kazuo Takayama, Kazutoshi Takahashi, Sayaka Deguchi, and Renxing Yi
- Subjects
Camostat ,Proteases ,ACE2 ,camostat ,RM1-950 ,Biology ,Cathepsin B ,human iPS cells ,chemistry.chemical_compound ,Drug Discovery ,Receptor ,TMPRSS2 ,Cathepsin ,Gene knockdown ,CA-074 methyl ester ,SARS-CoV-2 ,COVID-19 ,CRISPRi ,Molecular biology ,chemistry ,Basigin ,Molecular Medicine ,Original Article ,Therapeutics. Pharmacology ,Viral load - Abstract
It has been reported that many receptors and proteases are required for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. Although angiotensin-converting enzyme 2 (ACE2) is the most important of these receptors, little is known about the contribution of other genes. In this study, we examined the roles of neuropilin-1, basigin, transmembrane serine proteases (TMPRSSs), and cathepsins (CTSs) in SARS-CoV-2 infection using the CRISPR interference system and ACE2-expressing human induced pluripotent stem (iPS) cells. Double knockdown of TMPRSS2 and cathepsin B (CTSB) reduced the viral load to 0.036% ± 0.021%. Consistently, the combination of the CTPB inhibitor CA-074 methyl ester and the TMPRSS2 inhibitor camostat reduced the viral load to 0.0078% ± 0.0057%. This result was confirmed using four SARS-CoV-2 variants (B.1.3, B.1.1.7, B.1.351, and B.1.1.248). The simultaneous use of these two drugs reduced viral load to less than 0.01% in both female and male iPS cells. These findings suggest that compounds targeting TMPRSS2 and CTSB exhibit highly efficient antiviral effects independent of gender and SARS-CoV-2 variant., Graphical abstract, Hashimoto et al. used iPS cells and the CRISPRi system to compare the functions of receptors and proteases involved in SARS-CoV-2 infection. Dual inhibition of TMPRSS2 and CTSB prevented SARS-CoV-2 infection. The combination of TMPRSS2 and CTSB inhibitor treatment exhibited highly efficient antiviral effects independent of gender and SARS-CoV-2 variant.
- Published
- 2021
45. Air-liquid interphase culture confers SARS-CoV-2 susceptibility to A549 alveolar epithelial cells
- Author
-
Yasuko Orba, William W. Hall, Shinsuke Toba, Takao Sanaki, Kentaro Uemura, Hirofumi Sawa, Akihiko Sato, Yukari Itakura, Koshiro Tabata, Kittiya Intaruck, Michihito Sasaki, and Mai Kishimoto
- Subjects
Camostat ,viruses ,Biophysics ,Cell Culture Techniques ,ACE2 ,Biology ,Peptidyl-Dipeptidase A ,Biochemistry ,Article ,Microbiology ,chemistry.chemical_compound ,Downregulation and upregulation ,Viral entry ,Humans ,A549 cells ,Respiratory system ,Receptor ,Molecular Biology ,Cells, Cultured ,TMPRSS2 ,A549 cell ,SARS-CoV-2 ,Serine Endopeptidases ,COVID-19 ,Cell Biology ,respiratory system ,Mucus ,respiratory tract diseases ,Up-Regulation ,Air-liquid interface ,Gene Expression Regulation, Neoplastic ,chemistry ,Cell culture ,Alveolar Epithelial Cells ,Disease Susceptibility - Abstract
The human lung cell A549 is susceptible to infection with a number of respiratory viruses. However, A549 cells are resistant to Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) infection in conventional submerged culture, and this would appear to be due to low expression levels of the SARSCoV-2 entry receptor: angiotensin-converting enzyme-2 (ACE2). Here, we examined SARS-CoV-2 susceptibility to A549 cells after adaptation to air-liquid interface (ALI) culture. A549 cells in ALI culture yielded a layer of mucus on their apical surface, exhibited decreased expression levels of the proliferation marker KI-67 and intriguingly became susceptible to SARS-CoV-2 infection. We found that A549 cells increased the endogenous expression levels of ACE2 and TMPRSS2 following adaptation to ALI culture conditions. Camostat, a TMPRSS2 inhibitor, reduced SARS-CoV-2 infection in ALI-cultured A549 cells. These findings indicate that ALI culture switches the phenotype of A549 cells from resistance to susceptibility to SARS-CoV-2 infection through upregulation of ACE2 and TMPRSS2. (c) 2021 Elsevier Inc. All rights reserved.
- Published
- 2021
46. Improving the Inhibition of TMPRSS2 by Molecular Docking, to Decrease the Process Infection of SARS-CoV-2
- Author
-
J. L. Vique-Sánchez
- Subjects
Camostat ,Daclatasvir ,Darexaban ,Sivelestat ,Pharmacology ,Biochemistry ,Otamixaban ,Argatroban ,chemistry.chemical_compound ,Nafamostat ,chemistry ,Gabexate ,medicine ,Molecular Medicine ,Molecular Biology ,Biotechnology ,medicine.drug - Abstract
COVID-19 pandemic continues with several works focused on the repositioning of drugs, vaccines, and antibodies against COVID-19, as well as new therapeutic targets on the cellular membrane (ACE2, NRP1, and TMPRSS2) that interacting with SARS-CoV-2 S-protein. This study proposes ten compounds (T1-T10) selected by molecular docking using a library of nearly 500,000 compounds, these ten compounds have better interaction than Daclatasvir, Ombitasvir, Camostat, Edoxaban, NCGC00386477, Nafamostat, NCGC00386945, Otamixaban, Darexaban, Gabexate, Letaxaban, Argatroban, Sivelestat, NCGC00385043, and Bromhexine, and all of them have an inhibitory effect reported at TMPRSS2. The T1-T10 compounds were selected by molecular docking in the catalytic site of TMPRSS2, which could hinder/block the interaction with the S-protein and ACE2. Therefore the initial/early stage of COVID-19 could be avoided or decreased by hindering the fusion between SARS-CoV-2 and the cell membrane and this way to develop a new adjuvant treatment against COVID-19. © 2021 by the authors.
- Published
- 2021
- Full Text
- View/download PDF
47. Comprehensive Cardiotoxicity Assessment of COVID-19 Treatments Using Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes
- Author
-
Sayo Hayashi, Atsushi Ono, Ayano Satsuka, Yasunari Kanda, and Shota Yanagida
- Subjects
Camostat ,Induced Pluripotent Stem Cells ,030204 cardiovascular system & hematology ,Pharmacology ,Favipiravir ,Toxicology ,Afterdepolarization ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Humans ,Myocytes, Cardiac ,030212 general & internal medicine ,Induced pluripotent stem cell ,Cells, Cultured ,Cardiotoxicity ,SARS-CoV-2 ,business.industry ,Hydroxychloroquine ,COVID-19 Drug Treatment ,Clinical trial ,Drug repositioning ,chemistry ,business ,medicine.drug - Abstract
Coronavirus disease 2019 (COVID-19) continues to spread across the globe, with numerous clinical trials underway seeking to develop and test effective COVID-19 therapies, including remdesivir. Several ongoing studies have reported hydroxychloroquine-induced cardiotoxicity, including development of torsade de pointes (TdP). Meanwhile, human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are expected to serve as a tool for assessing drug-induced cardiotoxicity, such as TdP and contraction impairment. However, the cardiotoxicity of COVID-19 treatments has not been fully assessed using hiPSC-CMs. In this study, we focused on drug repurposing with various modes of actions and examined the TdP risk associated with COVID-19 treatments using field potential using multi-electrode array system and motion analysis with hiPSC-CMs. Hydroxychloroquine induced early after depolarization, while remdesivir, favipiravir, camostat, and ivermectin had little effect on field potentials. We then analyzed electromechanical window, which is defined as the difference between field potential and contraction-relaxation durations. Hydroxychloroquine decreased electromechanical window of hiPSC-CMs in a concentration-dependent manner. In contrast, other drugs had little effect. Our data suggest that hydroxychloroquine has proarrhythmic risk and other drugs have low proarrhythmic risk. Thus, hiPSC-CMs represent a useful tool for assessing the comprehensive cardiotoxicity caused by COVID-19 treatments in nonclinical settings.
- Published
- 2021
- Full Text
- View/download PDF
48. Cross-linking peptide and repurposed drugs inhibit both entry pathways of SARS-CoV-2
- Author
-
Kelvin K. W. To, Hoiyan Lam, Jasper Fuk-Woo Chan, Wan-Mui Chan, Chris Chung-Sing Chan, Jonathan Daniel Ip, Man Lung Yeung, Xinxin Zhou, Kwok-Yung Yuen, Hanjun Zhao, Shibo Jiang, Anna Jinxia Zhang, Jian-Piao Cai, Allen Wing-Ho Chu, Zheng Peng, and Andrew Chak-Yiu Lee
- Subjects
0301 basic medicine ,Indoles ,animal diseases ,viruses ,General Physics and Astronomy ,Pharmacology ,medicine.disease_cause ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Chloroquine ,Chlorocebus aethiops ,skin and connective tissue diseases ,Coronavirus ,Mice, Inbred BALB C ,Multidisciplinary ,Drug discovery ,Serine Endopeptidases ,virus diseases ,Drug repositioning ,030220 oncology & carcinogenesis ,Female ,medicine.drug ,Camostat ,Endosome ,Science ,Antiviral Agents ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,In vivo ,medicine ,Animals ,Humans ,Vero Cells ,SARS-CoV-2 ,HEK 293 cells ,fungi ,Drug Repositioning ,COVID-19 ,General Chemistry ,Virus Internalization ,COVID-19 Drug Treatment ,030104 developmental biology ,HEK293 Cells ,chemistry ,Peptides - Abstract
Up to date, effective antivirals have not been widely available for treating COVID-19. In this study, we identify a dual-functional cross-linking peptide 8P9R which can inhibit the two entry pathways (endocytic pathway and TMPRSS2-mediated surface pathway) of SARS-CoV-2 in cells. The endosomal acidification inhibitors (8P9R and chloroquine) can synergistically enhance the activity of arbidol, a spike-ACE2 fusion inhibitor, against SARS-CoV-2 and SARS-CoV in cells. In vivo studies indicate that 8P9R or the combination of repurposed drugs (umifenovir also known as arbidol, chloroquine and camostat which is a TMPRSS2 inhibitor), simultaneously interfering with the two entry pathways of coronaviruses, can significantly suppress SARS-CoV-2 replication in hamsters and SARS-CoV in mice. Here, we use drug combination (arbidol, chloroquine, and camostat) and a dual-functional 8P9R to demonstrate that blocking the two entry pathways of coronavirus can be a promising and achievable approach for inhibiting SARS-CoV-2 replication in vivo. Cocktail therapy of these drug combinations should be considered in treatment trials for COVID-19., Until today effective antivirals for COVID-19 treatment are not widely available. Here, Zhao et al. characterize a dual-functional cross-linking peptide, 8P9R, that can inhibit SARS-CoV-2 virus entry in vitro and suppresses viral replication in vivo in golden Syrian hamster.
- Published
- 2021
49. Pharmacoinformatics-based identification of transmembrane protease serine-2 inhibitors from Morus Alba as SARS-CoV-2 cell entry inhibitors
- Author
-
Siham A. Alissa, Hassna Mohammed Alhajri, Shuchi Nagar, Tahani Mazyad Almutairi, Rupesh V. Chikhale, Surajit Ghosh, Hans Raj Bhat, Ataul Islam, Fatmah A.S. Alasmary, and Anshul Shakya
- Subjects
Camostat ,Virtual screening ,medicine.medical_treatment ,030303 biophysics ,Molecular Dynamics Simulation ,medicine.disease_cause ,urologic and male genital diseases ,Catalysis ,Inorganic Chemistry ,Hydrophobic effect ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Discovery ,Morus alba Linn ,medicine ,Serine ,Humans ,Protease Inhibitors ,Physical and Theoretical Chemistry ,Molecular Biology ,TMPRSS2 ,030304 developmental biology ,Coronavirus ,chemistry.chemical_classification ,0303 health sciences ,Protease ,Chemistry ,SARS-CoV-2 ,Organic Chemistry ,General Medicine ,Virus Internalization ,Transmembrane Protease Serine 2 ,Transmembrane protein ,Amino acid ,COVID-19 Drug Treatment ,Molecular Docking Simulation ,Biochemistry ,Molecular docking ,Original Article ,Morus ,Information Systems - Abstract
Abstract Transmembrane protease serine-2 (TMPRSS2) is a cell-surface protein expressed by epithelial cells of specific tissues including those in the aerodigestive tract. It helps the entry of novel coronavirus (n-CoV) or Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) in the host cell. Successful inhibition of the TMPRSS2 can be one of the crucial strategies to stop the SARS-CoV-2 infection. In the present study, a set of bioactive molecules from Morus alba Linn. were screened against the TMPRSS2 through two widely used molecular docking engines such as Autodock vina and Glide. Molecules having a higher binding affinity toward the TMPRSS2 compared to Camostat and Ambroxol were considered for in-silico pharmacokinetic analyses. Based on acceptable pharmacokinetic parameters and drug-likeness, finally, five molecules were found to be important for the TMPRSS2 inhibition. A number of bonding interactions in terms of hydrogen bond and hydrophobic interactions were observed between the proposed molecules and ligand-interacting amino acids of the TMPRSS2. The dynamic behavior and stability of best-docked complex between TRMPRSS2 and proposed molecules were assessed through molecular dynamics (MD) simulation. Several parameters from MD simulation have suggested the stability between the protein and ligands. Binding free energy of each molecule calculated through MM-GBSA approach from the MD simulation trajectory suggested strong affection toward the TMPRSS2. Hence, proposed molecules might be crucial chemical components for the TMPRSS2 inhibition. Graphic abstract
- Published
- 2021
50. Strong Binding of Leupeptin with TMPRSS2 Protease May Be an Alternative to Camostat and Nafamostat for SARS-CoV-2 Repurposed Drug: Evaluation from Molecular Docking and Molecular Dynamics Simulations
- Author
-
Ramakrishnan, Jaganathan, Kandasamy, Saravanan, Iruthayaraj, Ancy, Magudeeswaran, Sivanandam, Chinnasamy, Kalaiarasi, and Poomani, Kumaradhas
- Subjects
Camostat ,Leupeptins ,medicine.medical_treatment ,Bioengineering ,Molecular dynamics ,Molecular Dynamics Simulation ,Pharmacology ,urologic and male genital diseases ,Antiviral Agents ,Guanidines ,Applied Microbiology and Biotechnology ,Biochemistry ,Molecular Docking Simulation ,Article ,chemistry.chemical_compound ,medicine ,Humans ,Molecular Biology ,TMPRSS2 ,Protease ,SARS-CoV-2 ,Chemistry ,Serine Endopeptidases ,Leupeptin ,Drug Repositioning ,COVID-19 ,Respiratory infection ,Esters ,Repurposed drug ,General Medicine ,Transmembrane Protease Serine 2 ,Benzamidines ,COVID-19 Drug Treatment ,Nafamostat ,Binding affinity ,Docking (molecular) ,Molecular docking ,Protein Binding ,Biotechnology - 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. Supplementary Information The online version contains supplementary material available at 10.1007/s12010-020-03475-8.
- 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.