15 research outputs found

    Synthesis of a thiazole library via an iridium-catalyzed sulfur ylide insertion reaction

    Get PDF
    A library of thiazoles and selenothiazoles were synthesized via Ir-catalyzed ylide insertion chemistry. This process is a functional group, particularly heterocycle-substituent tolerant. This was applied to the synthesis of fanetizole, an anti-inflammatory drug, and a thiazole-containing drug fragment that binds to the peptidyl-tRNA hydrolase (Pth) in Neisseria gonorrheae bacteria

    Accelerating drug target inhibitor discovery with a deep generative foundation model

    Get PDF
    Inhibitor discovery for emerging drug-target proteins is challenging, especially when target structure or active molecules are unknown. Here, we experimentally validate the broad utility of a deep generative framework trained at-scale on protein sequences, small molecules, and their mutual interactions-unbiased toward any specific target. We performed a protein sequence-conditioned sampling on the generative foundation model to design small-molecule inhibitors for two dissimilar targets: the spike protein receptor-binding domain (RBD) and the main protease from SARS-CoV-2. Despite using only the target sequence information during the model inference, micromolar-level inhibition was observed in vitro for two candidates out of four synthesized for each target. The most potent spike RBD inhibitor exhibited activity against several variants in live virus neutralization assays. These results establish that a single, broadly deployable generative foundation model for accelerated inhibitor discovery is effective and efficient, even in the absence of target structure or binder information

    Open science discovery of potent noncovalent SARS-CoV-2 main protease inhibitors

    Get PDF
    INTRODUCTION COVID-19 became a global pandemic partially as a result of the lack of easily deployable, broad-spectrum oral antivirals, which complicated its containment. Even endemically, and with effective vaccinations, it will continue to cause acute disease, death, and long-term sequelae globally unless there are accessible treatments. COVID-19 is not an isolated event but instead is the latest example of a viral pandemic threat to human health. Therefore, antiviral discovery and development should be a key pillar of pandemic preparedness efforts. RATIONALE One route to accelerate antiviral drug discovery is the establishment of open knowledge bases, the development of effective technology infrastructures, and the discovery of multiple potent antivirals suitable as starting points for the development of therapeutics. In this work, we report the results of the COVID Moonshot—a fully open science, crowdsourced, and structure-enabled drug discovery campaign—against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease (Mpro). This collaboration may serve as a roadmap for the potential development of future antivirals. RESULTS On the basis of the results of a crystallographic fragment screen, we crowdsourced design ideas to progress from fragment to lead compounds. The crowdsourcing strategy yielded several key compounds along the optimization trajectory, including the starting compound of what became the primary lead series. Three additional chemically distinct lead series were also explored, spanning a diversity of chemotypes. The collaborative and highly automated nature of the COVID Moonshot Consortium resulted in >18,000 compound designs, >2400 synthesized compounds, >490 ligand-bound x-ray structures, >22,000 alchemical free-energy calculations, and >10,000 biochemical measurements—all of which were made publicly available in real time. The recently approved antiviral ensitrelvir was identified in part based on crystallographic data from the COVID Moonshot Consortium. This campaign led to the discovery of a potent [median inhibitory concentration (IC50) = 37 ± 2 nM] and differentiated (noncovalent and nonpeptidic) lead compound that also exhibited potent cellular activity, with a median effective concentration (EC50) of 64 nM in A549-ACE2-TMPRSS2 cells and 126 nM in HeLa-ACE2 cells without measurable cytotoxicity. Although the pharmacokinetics of the reported compound is not yet optimal for therapeutic development, it is a promising starting point for further antiviral discovery and development. CONCLUSION The success of the COVID Moonshot project in producing potent antivirals, building open knowledge bases, accelerating external discovery efforts, and functioning as a useful information-exchange hub is an example of the potential effectiveness of open science antiviral discovery programs. The open science, patent-free nature of the project enabled a large number of collaborators to provide in-kind support, including synthesis, assays, and in vitro and in vivo experiments. By making all data immediately available and ensuring that all compounds are purchasable from Enamine without the need for materials transfer agreements, we aim to accelerate research globally along parallel tracks. In the process, we generated a detailed map of the structural plasticity of Mpro, extensive structure-activity relationships for multiple chemotypes, and a wealth of biochemical activity data to spur further research into antivirals and discovery methodologies. We hope that this can serve as an alternative model for antiviral discovery and future pandemic preparedness. Further, the project also showcases the role of machine learning, computational chemistry, and high-throughput structural biology as force multipliers in drug design. Artificial intelligence and machine learning algorithms help accelerate chemical synthesis while balancing multiple competing molecular properties. The design-make-test-analyze cycle was accelerated by these algorithms combined with planetary-scale biomolecular simulations of protein-ligand interactions and rapid structure determination

    Expanding the repertoire of low‐molecular‐weight pentafluorosulfanyl‐substituted scaffolds

    Get PDF
    The pentafluorosulfanyl (-SF5) functional group is of increasing interest as a bioisostere in medicinal chemistry. A library of SF5-containing compounds, including amide, isoxazole, and oxindole derivatives, was synthesised using a range of solution-based and solventless methods, including microwave and ball-mill techniques. The library was tested against targets including human dihydroorotate dehydrogenase (HDHODH). A subsequent focused approach led to synthesis of analogues of the clinically used disease modifying anti-rheumatic drugs (DMARDs), Teriflunomide and Leflunomide, considered for potential COVID-19 use, where SF5 bioisostere deployment led to improved inhibition of HDHODH compared with the parent drugs. The results demonstrate the utility of the SF5 group in medicinal chemistry

    A community effort in SARS-CoV-2 drug discovery.

    Get PDF
    peer reviewedThe COVID-19 pandemic continues to pose a substantial threat to human lives and is likely to do so for years to come. Despite the availability of vaccines, searching for efficient small-molecule drugs that are widely available, including in low- and middle-income countries, is an ongoing challenge. In this work, we report the results of an open science community effort, the "Billion molecules against Covid-19 challenge", to identify small-molecule inhibitors against SARS-CoV-2 or relevant human receptors. Participating teams used a wide variety of computational methods to screen a minimum of 1 billion virtual molecules against 6 protein targets. Overall, 31 teams participated, and they suggested a total of 639,024 molecules, which were subsequently ranked to find 'consensus compounds'. The organizing team coordinated with various contract research organizations (CROs) and collaborating institutions to synthesize and test 878 compounds for biological activity against proteases (Nsp5, Nsp3, TMPRSS2), nucleocapsid N, RdRP (only the Nsp12 domain), and (alpha) spike protein S. Overall, 27 compounds with weak inhibition/binding were experimentally identified by binding-, cleavage-, and/or viral suppression assays and are presented here. Open science approaches such as the one presented here contribute to the knowledge base of future drug discovery efforts in finding better SARS-CoV-2 treatments.R-AGR-3826 - COVID19-14715687-CovScreen (01/06/2020 - 31/01/2021) - GLAAB Enric

    Accelerating Inhibitor Discovery for Multiple SARS-CoV-2 Targets with a Single, Sequence-Guided Deep Generative Framework

    Full text link
    The COVID-19 pandemic has highlighted the urgency for developing more efficient molecular discovery pathways. As exhaustive exploration of the vast chemical space is infeasible, discovering novel inhibitor molecules for emerging drug-target proteins is challenging, particularly for targets with unknown structure or ligands. We demonstrate the broad utility of a single deep generative framework toward discovering novel drug-like inhibitor molecules against two distinct SARS-CoV-2 targets -- the main protease (Mpro) and the receptor binding domain (RBD) of the spike protein. To perform target-aware design, the framework employs a target sequence-conditioned sampling of novel molecules from a generative model. Micromolar-level in vitro inhibition was observed for two candidates (out of four synthesized) for each target. The most potent spike RBD inhibitor also emerged as a rare non-covalent antiviral with broad-spectrum activity against several SARS-CoV-2 variants in live virus neutralization assays. These results show that a broadly deployable machine intelligence framework can accelerate hit discovery across different emerging drug-targets.Comment: Fixed dates, added data availability, minor change

    Iterative computational design and crystallographic screening identifies potent inhibitors targeting the Nsp3 macrodomain of SARS-CoV-2.

    No full text
    The nonstructural protein 3 (NSP3) of the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) contains a conserved macrodomain enzyme (Mac1) that is critical for pathogenesis and lethality. While small-molecule inhibitors of Mac1 have great therapeutic potential, at the outset of the COVID-19 pandemic, there were no well-validated inhibitors for this protein nor, indeed, the macrodomain enzyme family, making this target a pharmacological orphan. Here, we report the structure-based discovery and development of several different chemical scaffolds exhibiting low- to sub-micromolar affinity for Mac1 through iterations of computer-aided design, structural characterization by ultra-high-resolution protein crystallography, and binding evaluation. Potent scaffolds were designed with in silico fragment linkage and by ultra-large library docking of over 450 million molecules. Both techniques leverage the computational exploration of tangible chemical space and are applicable to other pharmacological orphans. Overall, 160 ligands in 119 different scaffolds were discovered, and 153 Mac1-ligand complex crystal structures were determined, typically to 1 Å resolution or better. Our analyses discovered selective and cell-permeable molecules, unexpected ligand-mediated conformational changes within the active site, and key inhibitor motifs that will template future drug development against Mac1

    Allosteric regulation and crystallographic fragment screening of SARS-CoV-2 NSP15 endoribonuclease

    No full text
    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19). The NSP15 endoribonuclease enzyme, known as NendoU, is highly conserved and plays a critical role in the ability of the virus to evade the immune system. NendoU is a promising target for the development of new antiviral drugs. However, the complexity of the enzyme's structure and kinetics, along with the broad range of recognition sequences and lack of structural complexes, hampers the development of inhibitors. Here, we performed enzymatic characterization of NendoU in its monomeric and hexameric form, showing that hexamers are allosteric enzymes with a positive cooperative index, and with no influence of manganese on enzymatic activity. Through combining cryo-electron microscopy at different pHs, X-ray crystallography and biochemical and structural analysis, we showed that NendoU can shift between open and closed forms, which probably correspond to active and inactive states, respectively. We also explored the possibility of NendoU assembling into larger supramolecular structures and proposed a mechanism for allosteric regulation. In addition, we conducted a large fragment screening campaign against NendoU and identified several new allosteric sites that could be targeted for the development of new inhibitors. Overall, our findings provide insights into the complex structure and function of NendoU and offer new opportunities for the development of inhibitors

    Expanding the repertoire of low-molecular-weight pentafluorosulfanyl-substituted scaffolds

    No full text
    The pentafluorosulfanyl (-SF5) functional group is of increasing interest as a bioisostere in medicinal chemistry. A library of SF5-containing compounds, including amide, isoxazole, and oxindole derivatives, was synthesised using a range of solution-based and solventless methods, including microwave and ball-mill techniques. The library was tested against targets including human dihydroorotate dehydrogenase (HDHODH). A subsequent focused approach led to synthesis of analogues of the clinically used disease modifying anti-rheumatic drugs (DMARDs), Teriflunomide and Leflunomide, considered for potential COVID-19 use, where SF5 bioisostere deployment led to improved inhibition of HDHODH compared with the parent drugs. The results demonstrate the utility of the SF5 group in medicinal chemistry
    corecore