9 research outputs found

    Investigation into the Use of Encorafenib to Develop Potential PROTACs Directed against BRAFV600E Protein

    Get PDF
    BRAF is a serine/threonine kinase frequently mutated in human cancers. BRAF(V600E) mutated protein is targeted through the use of kinase inhibitors which are approved for the treatment of melanoma; however, their long-term efficacy is hampered by resistance mechanisms. The PROTAC-induced degradation of BRAF(V600E) has been proposed as an alternative strategy to avoid the onset of resistance. In this study, we designed a series of compounds where the BRAF kinase inhibitor encorafenib was conjugated to pomalidomide through different linkers. The synthesized compounds maintained their ability to inhibit the kinase activity of mutated BRAF with IC(50) values in the 40–88 nM range. Selected compounds inhibited BRAF(V600E) signaling and cellular proliferation of A375 and Colo205 tumor cell lines. Compounds 10 and 11, the most active of the series, were not able to induce degradation of mutated BRAF. Docking and molecular dynamic studies, conducted in comparison with the efficient BRAF degrader P5B, suggest that a different orientation of the linker bearing the pomalidomide substructure, together with a decreased mobility of the solvent-exposed part of the conjugates, could explain this behavior

    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

    Inhibition of Patched chemotherapy resistance activity : biocomputational, chemical and cellular approaches

    No full text
    MalgrĂ© des efforts continus dans le dĂ©veloppement de nouveaux mĂ©dicaments, la rĂ©sistance aux chimiothĂ©rapies reste un dĂ©fi majeur dans le traitement du cancer. L'un des principaux mĂ©canismes responsables de la rĂ©sistance aux mĂ©dicaments est l'efflux des agents thĂ©rapeutiques hors des cellules cancĂ©reuses par les transporteurs multidrogues.Le rĂ©cepteur Hedgehog Patched1 (PTCH1), qui fait partie de la voie de signalisation Hedgehog, est surexprimĂ© dans de nombreux cancers. Outre son rĂŽle physiologique de transporteur de cholestĂ©rol, PTCH1 est aussi capable de transporter des agents anticancĂ©reux hors des cellules cancĂ©reuses, contribuant ainsi Ă  la rĂ©sistance aux mĂ©dicaments avec les transporteurs ABC (ATP binding cassette). Contrairement Ă  ces derniers, qui sont exprimĂ©s de maniĂšre endogĂšne dans les cellules normales et jouent un rĂŽle crucial dans la survie cellulaire, l'activitĂ© d'efflux de PTCH1 n'a lieu que dans les cellules cancĂ©reuses. Cela fait de PTCH1 une nouvelle cible prometteuse pour le traitement du cancer. Trois inhibiteurs de son activitĂ© d'efflux ont Ă©tĂ© identifiĂ©s Ă  ce jour : l'astĂ©mizole, la mĂ©thiothĂ©pine et un composĂ© naturel, la panicĂ©ine A hydroquinone (PAH). Ces composĂ©s augmentent l'efficacitĂ© des chimiothĂ©rapies contre les cellules de mĂ©lanome in vitro et in vivo, prouvant que l'inhibition de PTCH1 est une stratĂ©gie anticancĂ©reuse pertinente. Cependant, ils prĂ©sentent des limites intrinsĂšques et doivent ĂȘtre optimisĂ©s. Par exemple, la PAH a une trĂšs faible stabilitĂ© mĂ©tabolique, ce qui empĂȘche d'envisager son dĂ©veloppement en tant que mĂ©dicament.Dans ce contexte, le projet a pour objectif de dĂ©velopper un inhibiteur de l'efflux de PTCH1, en tant que mĂ©dicament “first-in-class” pour lutter contre la rĂ©sistance Ă  la chimiothĂ©rapie et amĂ©liorer la survie des patients grĂące Ă  une optimisation rationnelle de la PAH.Nous avons donc Ă©tudiĂ© l'interaction entre la PAH et PTCH1 au moyen de mĂ©thodologies in silico et effectuĂ© une comparaison structure-ligand entre les trois inhibiteurs connus. En utilisant un protocole de docking couplĂ© Ă  des simulations de dynamiques molĂ©culaires, nous avons extrait des informations importantes sur la conformation active d'un inhibiteur de PTCH1 et identifiĂ© un site de liaison supposĂ© dans le canal hydrophobe de PTCH1.Nous avons mis au point une nouvelle synthĂšse totale de l'E-PAH qui rĂ©pond Ă  certaines limitations importantes de la prĂ©cĂ©dente, telles que sa non-stĂ©rĂ©osĂ©lectivitĂ© et sa grande spĂ©cificitĂ© de substrat. AprĂšs optimisation, nous avons finalement obtenu un nouveau protocole de synthĂšse stĂ©rĂ©osĂ©lective et robuste pour les E et Z PAH. Ceci a permis de synthĂ©tiser plusieurs analogues de PAH dont l'activitĂ© a Ă©tĂ© Ă©valuĂ©e sur des cellules de mĂ©lanome, en combinaison avec le vemurafenib, dans le but de rĂ©aliser une Ă©tude structure-activitĂ© pour le motif hydroquinone. Nous avons Ă©galement Ă©valuĂ© certaines propriĂ©tĂ©s pharmacocinĂ©tiques de ce motif, telles que la stabilitĂ© plasmatique et le profil de sĂ©curitĂ©.La partie hydroquinone de la PAH est importante pour son activitĂ© biologique, mais c'est aussi l'un de ses principaux inconvĂ©nients en raison de son oxydation facile en quinone. En combinant les connaissances acquises grĂące aux Ă©tudes in silico et SAR dans la derniĂšre partie de ce projet, nous avons cherchĂ© Ă  synthĂ©tiser des nouvelles molĂ©cules en appliquant un remplacement "bioisostĂ©rique", une approche souvent utilisĂ©e avec succĂšs en chimie mĂ©dicinale. Tout d'abord, le remplacement de la double liaison de la PAH par des groupes chimiques biologiquement Ă©quivalents a permis une stratĂ©gie de synthĂšse robuste, convergente et efficace, qui s'inscrit dans les tendances modernes des approches de chimie durable. Ensuite, il a Ă©tĂ© possible de synthĂ©tiser facilement un grand nombre de composĂ©s, en remplaçant le motif hydroxyquinone, et de cribler l'activitĂ© biologique in cellulo d'un espace chimique diversifiĂ© avec des rĂ©sultats biologiques trĂšs prometteurs.Despite the continuous efforts in the design and development of new drugs with innovative mode of action, resistance to both chemo- and targeted therapy remains a major challenge in cancer treatment. One of the major mechanisms responsible for multidrug resistance is the efflux of therapeutics out of cancer cells by multidrug transporters.The Hedgehog receptor Patched1 (PTCH1), part of the Hedgehog signaling pathway, is over-expressed in many cancers. In addition to its physiological role as cholesterol transporter, PTCH1 is also able to transport anticancer agents out of cancer cells, thus contributing to multidrug resistance together with the ATP binding cassette (ABC) transporters. Unlike ABC transporters which are endogenously expressed in normal cells and play a crucial role in cell survival, PTCH1 efflux activity takes place only in cancer cells. This makes PTCH1 a new attractive target for cancer treatment. Three inhibitors of PTCH1 efflux activity have been identified to date, namely, astemizole, methiothepin and the natural compound panicein A hydroquinone (PAH). These compounds increased the efficacy of both conventional and targeted chemotherapies against melanoma cells in vitro and in vivo, showing the proof of concept of PTCH1 inhibition as a successful anticancer strategy. However, they have some intrinsic limitations and require optimization. For instance, PAH, has a very low metabolic stability which prevents its advance in a drug development pipeline towards the clinics.In that context, this PhD project is aimed at the ultimate goal of developing a PTCH1 drug efflux inhibitor as a first-in-class drug candidate to fight chemotherapy resistance and improve patient survival. In particular, it comprehends the medicinal chemistry efforts made in the direction of rational optimization of the natural compound PAH.To this aim we studied the interaction between PAH and PTCH1 by means of in silico methodologies and we performed structure- and ligand based comparison between the three known inhibitors. By using an ensemble docking protocol coupled to molecular dynamics simulations of the ligands we extracted important information about the active conformation of a PTCH1 inhibitor and we identified a putative binding site within the hydrophobic channel on PTCH1.We developed a new total synthesis for E-PAH which addresses some important limitations of the previous one, such as its non-stereoselectivity and high substrate specificity. We applied several strategies and eventually obtained a stereoselective and robust new synthesis protocol for E and Z PAH. This allowed for the synthesis of several PAH analogs whose activity was evaluated on melanoma cells, in combination with vemurafenib, with the aim to assess a structure-activity relationship for the hydroquinone scaffold. We further evaluated some PK properties of the hydroquinone scaffold such as plasma stability and safety profile.The hydroquinone moiety of PAH is important for its biological activity but it is also one of its major drawbacks due to its easy oxidation to quinone. By combining the knowledge acquired from the in silico and SAR studies on the hydroquinone, in the last part of this PhD project we aimed at the synthesis of innovative scaffolds by applying a “bioisosteric” replacement on the hydroquinone, an approach often successfully used in medicinal chemistry. First, the replacement of the double bond of PAH with biologically equivalent chemical groups allowed for a robust, convergent and efficient synthetic strategy, which is line with the modern trends of sustainable chemistry approaches. Next, it made possible to readily synthesize a large number of compounds, substituting the hydroxyquinone motif, and screen the in cellulo biological activity of a diversified chemical space with quite promising biological results

    Chasing new targets to fight antimicrobial resistance: identification of small molecules affecting the interaction of human hemoglobin with the IsdB hemophore of Staphylococcus aureus

    No full text
    Human hemoglobin (Hb) is the preferred iron source of Staphylococcus aureus. This pathogenic bacterium exploits a sophisticated protein machinery called Iron-regulated surface determinant (Isd) system to bind Hb, extract and internalize heme and finally degrade it to complete iron acquisition. IsdB, the surface exposed Hb receptor, is a proven virulence factor of S. aureus and the inhibition of its interaction with Hb can be pursued as a strategy to develop new classes of antimicrobials. To identify small molecules able to disrupt IsdB:Hb protein-protein interactions (PPIs), we carried out a structure-based virtual screening campaign and developed an ad-hoc immunoassay to screen the retrieved set of commercially available compounds. Saturation-transfer difference (STD) NMR was applied to verify specific interactions of a sub-set of molecules, chosen based on their efficacy in reducing the amount of Hb bound to IsdB. Among molecules for which direct binding was verified, the best hit was submitted to ITC analysis to measure the binding affinity to Hb, which was found to be in the sub-micromolar range. The results demonstrate the viability of the proposed in silico/in vitro experimental pipeline to discover and test IsdB:Hb PPI inhibitors. The identified lead compound will be the starting point for future SAR and molecule optimization campaigns

    A community effort to discover small molecule SARS-CoV-2 inhibitors

    No full text
    The 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 a 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 potentially active 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 activity against proteases (Nsp5, Nsp3, TMPRSS2), nucleocapsid N, RdRP (Nsp12 domain), and (alpha) spike protein S. Overall, 27 potential inhibitors were experimentally confirmed by binding-, cleavage-, and/or viral suppression assays and are presented here. All results are freely available and can be taken further downstream without IP restrictions. Overall, we show the effectiveness of computational techniques, community efforts, and communication across research fields (i.e., protein expression and crystallography, in silico modeling, synthesis and biological assays) to accelerate the early phases of drug discovery
    corecore