scholarly journals Synthesis and Identification of Novel Potential Molecules Against COVID-19 Main Protease Through Structure-Guided Virtual Screening Approach

Author(s):  
Youness El Bakri ◽  
El Hassane Anouar ◽  
Sajjad Ahmad ◽  
Amal A. Nassar ◽  
Mohamed Labd Taha ◽  
...  
2020 ◽  
Vol 117 (44) ◽  
pp. 27381-27387 ◽  
Author(s):  
Zhe Li ◽  
Xin Li ◽  
Yi-You Huang ◽  
Yaoxing Wu ◽  
Runduo Liu ◽  
...  

The COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global crisis. There is no therapeutic treatment specific for COVID-19. It is highly desirable to identify potential antiviral agents against SARS-CoV-2 from existing drugs available for other diseases and thus repurpose them for treatment of COVID-19. In general, a drug repurposing effort for treatment of a new disease, such as COVID-19, usually starts from a virtual screening of existing drugs, followed by experimental validation, but the actual hit rate is generally rather low with traditional computational methods. Here we report a virtual screening approach with accelerated free energy perturbation-based absolute binding free energy (FEP-ABFE) predictions and its use in identifying drugs targeting SARS-CoV-2 main protease (Mpro). The accurate FEP-ABFE predictions were based on the use of a restraint energy distribution (RED) function, making the practical FEP-ABFE−based virtual screening of the existing drug library possible. As a result, out of 25 drugs predicted, 15 were confirmed as potent inhibitors of SARS-CoV-2 Mpro. The most potent one is dipyridamole (inhibitory constant Ki= 0.04 µM) which has shown promising therapeutic effects in subsequently conducted clinical studies for treatment of patients with COVID-19. Additionally, hydroxychloroquine (Ki= 0.36 µM) and chloroquine (Ki= 0.56 µM) were also found to potently inhibit SARS-CoV-2 Mpro. We anticipate that the FEP-ABFE prediction-based virtual screening approach will be useful in many other drug repurposing or discovery efforts.


Author(s):  
Zhe Li ◽  
Xin Li ◽  
Yi-You Huang ◽  
Yaoxing Wu ◽  
Runduo Liu ◽  
...  

AbstractCoronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global crisis. There is no therapeutic treatment specific for COVID-19. It is highly desirable to identify potential antiviral agents against SARS-CoV-2 from existing drugs available for other diseases and, thus, repurpose them for treatment of COVID-19. In general, a drug repurposing effort for treatment of a new disease, such as COVID-19, usually starts from a virtual screening of existing drugs, followed by experimental validation, but the actual hit rate is generally rather low with traditional computational methods. Here we report a new virtual screening approach with accelerated free energy perturbation-based absolute binding free energy (FEP-ABFE) predictions and its use in identifying drugs targeting SARS-CoV-2 main protease (Mpro). The accurate FEP-ABFE predictions were based on the use of a new restraint energy distribution (RED) function designed to accelerate the FEP-ABFE calculations and make the practical FEP-ABFE-based virtual screening of the existing drug library possible for the first time. As a result, out of twenty-five drugs predicted, fifteen were confirmed as potent inhibitors of SARS-CoV-2 Mpro. The most potent one is dipyridamole (Ki=0.04 μM) which has showed promising therapeutic effects in subsequently conducted clinical studies for treatment of patients with COVID-19. Additionally, hydroxychloroquine (Ki=0.36 μM) and chloroquine (Ki=0.56 μM) were also found to potently inhibit SARS-CoV-2 Mpro for the first time. We anticipate that the FEP-ABFE prediction-based virtual screening approach will be useful in many other drug repurposing or discovery efforts.Significance StatementDrug repurposing effort for treatment of a new disease, such as COVID-19, usually starts from a virtual screening of existing drugs, followed by experimental validation, but the actual hit rate is generally rather low with traditional computational methods. It has been demonstrated that a new virtual screening approach with accelerated free energy perturbation-based absolute binding free energy (FEP-ABFE) predictions can reach an unprecedently high hit rate, leading to successful identification of 16 potent inhibitors of SARS-CoV-2 main protease (Mpro) from computationally selected 25 drugs under a threshold of Ki = 4 μM. The outcomes of this study are valuable for not only drug repurposing to treat COVID-19, but also demonstrating the promising potential of the FEP-ABFE prediction-based virtual screening approach.


2020 ◽  
Author(s):  
Son Tung Ngo ◽  
Hung Minh Nguyen ◽  
Le Thi Thuy Huong ◽  
Pham Minh Quan ◽  
Vi Khanh Truong ◽  
...  

A virtual screening approach using docking and free energy pertubation was successfully validated with previously characterized inhibitors of SARS-CoV-2 main protease (Mpro). This approach and then used to estimate the binding affinity to Mpro of more than 6300 compounds in the ZINC15 database. Delamanid, an anti-tuberculosis agent, has a predicted nanomolar binding affinity for SARS-CoV-2 Mpro and is thus a promissing drug candiate for COVID-19. In addition, several compounds including three antibiotics exhibits femtomolar affinity for SARS-CoV-2 Mpro. The residues around positions 24, 45, 143, 165, and 190 were found to be involved in the binding of the strongest inhibitors.


RSC Advances ◽  
2015 ◽  
Vol 5 (30) ◽  
pp. 23202-23209 ◽  
Author(s):  
Ruijuan Li ◽  
Xiaolin Su ◽  
Zheng Chen ◽  
Wanxu Huang ◽  
Yali Wang ◽  
...  

Novel PAK4 inhibitors were discovered using structure-based virtual screening approach for further chemical modification.


2006 ◽  
Vol 19 (12) ◽  
pp. 1595-1601 ◽  
Author(s):  
Ching Y. Wang ◽  
Ni Ai ◽  
Sonia Arora ◽  
Karthigeyan Nagarajan ◽  
Randy Zauhar ◽  
...  

2011 ◽  
Vol 78 (6) ◽  
pp. 913-922 ◽  
Author(s):  
Alberto Massarotti ◽  
Sewan Theeramunkong ◽  
Ornella Mesenzani ◽  
Antonio Caldarelli ◽  
Armando A. Genazzani ◽  
...  

ChemInform ◽  
2007 ◽  
Vol 38 (42) ◽  
Author(s):  
Jian-Zhong Chen ◽  
Junmei Wang ◽  
Xiang-Qun Xie

Sign in / Sign up

Export Citation Format

Share Document