Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Nature ; 615(7954): 913-919, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36922589

RESUMO

Chromatin-binding proteins are critical regulators of cell state in haematopoiesis1,2. Acute leukaemias driven by rearrangement of the mixed lineage leukaemia 1 gene (KMT2Ar) or mutation of the nucleophosmin gene (NPM1) require the chromatin adapter protein menin, encoded by the MEN1 gene, to sustain aberrant leukaemogenic gene expression programs3-5. In a phase 1 first-in-human clinical trial, the menin inhibitor revumenib, which is designed to disrupt the menin-MLL1 interaction, induced clinical responses in patients with leukaemia with KMT2Ar or mutated NPM1 (ref. 6). Here we identified somatic mutations in MEN1 at the revumenib-menin interface in patients with acquired resistance to menin inhibition. Consistent with the genetic data in patients, inhibitor-menin interface mutations represent a conserved mechanism of therapeutic resistance in xenograft models and in an unbiased base-editor screen. These mutants attenuate drug-target binding by generating structural perturbations that impact small-molecule binding but not the interaction with the natural ligand MLL1, and prevent inhibitor-induced eviction of menin and MLL1 from chromatin. To our knowledge, this study is the first to demonstrate that a chromatin-targeting therapeutic drug exerts sufficient selection pressure in patients to drive the evolution of escape mutants that lead to sustained chromatin occupancy, suggesting a common mechanism of therapeutic resistance.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Leucemia , Mutação , Proteínas Proto-Oncogênicas , Animais , Humanos , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/genética , Cromatina/genética , Cromatina/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Leucemia/tratamento farmacológico , Leucemia/genética , Leucemia/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo
2.
J Chem Inf Model ; 64(10): 4009-4020, 2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38751014

RESUMO

Drug discovery pipelines nowadays rely on machine learning models to explore and evaluate large chemical spaces. While including 3D structural information is considered beneficial, structural models are hindered by the availability of protein-ligand complex structures. Exemplified for kinase drug discovery, we address this issue by generating kinase-ligand complex data using template docking for the kinase compound subset of available ChEMBL assay data. To evaluate the benefit of the created complex data, we use it to train a structure-based E(3)-invariant graph neural network. Our evaluation shows that binding affinities can be predicted with significantly higher precision by models that take synthetic binding poses into account compared to ligand- or drug-target interaction models alone.


Assuntos
Aprendizado de Máquina , Simulação de Acoplamento Molecular , Ligantes , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/metabolismo , Redes Neurais de Computação , Proteínas Quinases/metabolismo , Proteínas Quinases/química , Descoberta de Drogas/métodos , Ligação Proteica , Conformação Proteica , Fosfotransferases/metabolismo , Fosfotransferases/química , Fosfotransferases/antagonistas & inibidores
3.
Nucleic Acids Res ; 50(W1): W753-W760, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35524571

RESUMO

Computational pipelines have become a crucial part of modern drug discovery campaigns. Setting up and maintaining such pipelines, however, can be challenging and time-consuming-especially for novice scientists in this domain. TeachOpenCADD is a platform that aims to teach domain-specific skills and to provide pipeline templates as starting points for research projects. We offer Python-based solutions for common tasks in cheminformatics and structural bioinformatics in the form of Jupyter notebooks, based on open source resources only. Including the 12 newly released additions, TeachOpenCADD now contains 22 notebooks that cover both theoretical background as well as hands-on programming. To promote reproducible and reusable research, we apply software best practices to our notebooks such as testing with automated continuous integration and adhering to the idiomatic Python style. The new TeachOpenCADD website is available at https://projects.volkamerlab.org/teachopencadd and all code is deposited on GitHub.


Assuntos
Quimioinformática , Software , Biologia Computacional , Descoberta de Drogas
4.
J Chem Inf Model ; 62(10): 2600-2616, 2022 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-35536589

RESUMO

Protein kinases are among the most important drug targets because their dysregulation can cause cancer, inflammatory and degenerative diseases, and many more. Developing selective inhibitors is challenging due to the highly conserved binding sites across the roughly 500 human kinases. Thus, detecting subtle similarities on a structural level can help explain and predict off-targets among the kinase family. Here, we present the kinase-focused, subpocket-enhanced KiSSim fingerprint (Kinase Structural Similarity). The fingerprint builds on the KLIFS pocket definition, composed of 85 residues aligned across all available protein kinase structures, which enables residue-by-residue comparison without a computationally expensive alignment. The residues' physicochemical and spatial properties are encoded within their structural context including key subpockets at the hinge region, the DFG motif, and the front pocket. Since structure was found to contain information complementary to sequence, we used the fingerprint to calculate all-against-all similarities within the structurally covered kinome. We could identify off-targets that are unexpected if solely considering the sequence-based kinome tree grouping; for example, Erlobinib's known kinase off-targets SLK and LOK show high similarities to the key target EGFR (TK group), although belonging to the STE group. KiSSim reflects profiling data better or at least as well as other approaches such as KLIFS pocket sequence identity, KLIFS interaction fingerprints (IFPs), or SiteAlign. To rationalize observed (dis)similarities, the fingerprint values can be visualized in 3D by coloring structures with residue and feature resolution. We believe that the KiSSim fingerprint is a valuable addition to the kinase research toolbox to guide off-target and polypharmacology prediction. The method is distributed as an open-source Python package on GitHub and as a conda package: https://github.com/volkamerlab/kissim.


Assuntos
Inibidores de Proteínas Quinases , Proteínas Quinases , Sítios de Ligação , Humanos , Ligantes , Polifarmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo
5.
Biochem J ; 478(1): 217-234, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33241844

RESUMO

Smyd1 is an epigenetic modulator of gene expression that has been well-characterized in muscle cells. It was recently reported that Smyd1 levels are modulated by inflammatory processes. Since inflammation affects the vascular endothelium, this study aimed to characterize Smyd1 expression in endothelial cells. We detected Smyd1 in human endothelial cells (HUVEC and EA.hy926 cells), where the protein was largely localized in PML nuclear bodies (PML-NBs). By transfection of EA.hy926 cells with expression vectors encoding Smyd1, PML, SUMO1, active or mutant forms of the SUMO protease SuPr1 and/or the SUMO-conjugation enzyme UBC9, as well as Smyd1- or PML-specific siRNAs, in the presence or absence of the translation blocker cycloheximide or the proteasome-inhibitor MG132, and supported by computational modeling, we show that Smyd1 is SUMOylated in a PML-dependent manner and thereby addressed for degradation in proteasomes. Furthermore, transfection with Smyd1-encoding vectors led to PML up-regulation at the mRNA level, while PML transfection lowered Smyd1 protein stability. Incubation of EA.hy926 cells with the pro-inflammatory cytokine TNF-α resulted in a constant increase in Smyd1 mRNA and protein over 24 h, while incubation with IFN-γ induced a transient increase in Smyd1 expression, which peaked at 6 h and decreased to control values within 24 h. The IFN-γ-induced increase in Smyd1 was accompanied by more Smyd1 SUMOylation and more/larger PML-NBs. In conclusion, our data indicate that in endothelial cells, Smyd1 levels are regulated through a negative feedback mechanism based on SUMOylation and PML availability. This molecular control loop is stimulated by various cytokines.


Assuntos
Citocinas/farmacologia , Proteínas de Ligação a DNA/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Proteínas Musculares/metabolismo , Proteína da Leucemia Promielocítica/metabolismo , Sumoilação/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Núcleo Celular/metabolismo , Cicloeximida/farmacologia , Proteínas de Ligação a DNA/genética , Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Interferon gama/farmacologia , Leupeptinas/farmacologia , Proteínas Musculares/genética , Proteína da Leucemia Promielocítica/genética , Inibidores de Proteassoma/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/genética , RNA Interferente Pequeno , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Sumoilação/genética , Fatores de Transcrição/genética , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima
6.
J Chem Inf Model ; 61(7): 3255-3272, 2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34153183

RESUMO

Computational methods such as machine learning approaches have a strong track record of success in predicting the outcomes of in vitro assays. In contrast, their ability to predict in vivo endpoints is more limited due to the high number of parameters and processes that may influence the outcome. Recent studies have shown that the combination of chemical and biological data can yield better models for in vivo endpoints. The ChemBioSim approach presented in this work aims to enhance the performance of conformal prediction models for in vivo endpoints by combining chemical information with (predicted) bioactivity assay outcomes. Three in vivo toxicological endpoints, capturing genotoxic (MNT), hepatic (DILI), and cardiological (DICC) issues, were selected for this study due to their high relevance for the registration and authorization of new compounds. Since the sparsity of available biological assay data is challenging for predictive modeling, predicted bioactivity descriptors were introduced instead. Thus, a machine learning model for each of the 373 collected biological assays was trained and applied on the compounds of the in vivo toxicity data sets. Besides the chemical descriptors (molecular fingerprints and physicochemical properties), these predicted bioactivities served as descriptors for the models of the three in vivo endpoints. For this study, a workflow based on a conformal prediction framework (a method for confidence estimation) built on random forest models was developed. Furthermore, the most relevant chemical and bioactivity descriptors for each in vivo endpoint were preselected with lasso models. The incorporation of bioactivity descriptors increased the mean F1 scores of the MNT model from 0.61 to 0.70 and for the DICC model from 0.72 to 0.82 while the mean efficiencies increased by roughly 0.10 for both endpoints. In contrast, for the DILI endpoint, no significant improvement in model performance was observed. Besides pure performance improvements, an analysis of the most important bioactivity features allowed detection of novel and less intuitive relationships between the predicted biological assay outcomes used as descriptors and the in vivo endpoints. This study presents how the prediction of in vivo toxicity endpoints can be improved by the incorporation of biological information-which is not necessarily captured by chemical descriptors-in an automated workflow without the need for adding experimental workload for the generation of bioactivity descriptors as predicted outcomes of bioactivity assays were utilized. All bioactivity CP models for deriving the predicted bioactivities, as well as the in vivo toxicity CP models, can be freely downloaded from https://doi.org/10.5281/zenodo.4761225.


Assuntos
Fígado , Aprendizado de Máquina , Bioensaio , Conformação Molecular
7.
Arch Pharm (Weinheim) ; 354(9): e2100123, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34008218

RESUMO

The bioactive components of Garcinia indica, garcinol (camboginol), and isogarcinol (cambogin), are suitable drug candidates for the treatment of various human diseases. HIV-1-RNase H assay was used to study the RNase H inhibition by garcinol and isogarcinol. Docking of garcinol into the active site of the enzyme was carried out to rationalize the difference in activities between the two compounds. Garcinol showed higher HIV-1-RNase H inhibition than the known inhibitor RDS1759 and retained full potency against the RNase H of a drug-resistant HIV-1 reverse transcriptase form. Isogarcinol was distinctly less active than garcinol, indicating the importance of the enolizable ß-diketone moiety of garcinol for anti-RNase H activity. Docking calculations confirmed these findings and suggested this moiety to be involved in the chelation of metal ions of the active site. On the basis of its HIV-1 reverse transcriptase-associated RNase H inhibitory activity, garcinol is worth being further explored concerning its potential as a cost-effective treatment for HIV patients.


Assuntos
Garcinia/química , Inibidores da Transcriptase Reversa/farmacologia , Ribonuclease H do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Terpenos/farmacologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , HIV-1/enzimologia , Simulação de Acoplamento Molecular , Inibidores da Transcriptase Reversa/isolamento & purificação , Terpenos/isolamento & purificação
8.
Int J Mol Sci ; 22(9)2021 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-33922714

RESUMO

Drug discovery is a cost and time-intensive process that is often assisted by computational methods, such as virtual screening, to speed up and guide the design of new compounds. For many years, machine learning methods have been successfully applied in the context of computer-aided drug discovery. Recently, thanks to the rise of novel technologies as well as the increasing amount of available chemical and bioactivity data, deep learning has gained a tremendous impact in rational active compound discovery. Herein, recent applications and developments of machine learning, with a focus on deep learning, in virtual screening for active compound design are reviewed. This includes introducing different compound and protein encodings, deep learning techniques as well as frequently used bioactivity and benchmark data sets for model training and testing. Finally, the present state-of-the-art, including the current challenges and emerging problems, are examined and discussed.


Assuntos
Biologia Computacional/métodos , Aprendizado Profundo , Desenho de Fármacos , Descoberta de Drogas/métodos , Redes Neurais de Computação , Proteínas/química , Humanos , Tecnologia Farmacêutica
9.
Int J Mol Sci ; 22(5)2021 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-33668139

RESUMO

New 2-(thien-2-yl)-acrylonitriles with putative kinase inhibitory activity were prepared and tested for their antineoplastic efficacy in hepatoma models. Four out of the 14 derivatives were shown to inhibit hepatoma cell proliferation at (sub-)micromolar concentrations with IC50 values below that of the clinically relevant multikinase inhibitor sorafenib, which served as a reference. Colony formation assays as well as primary in vivo examinations of hepatoma tumors grown on the chorioallantoic membrane of fertilized chicken eggs (CAM assay) confirmed the excellent antineoplastic efficacy of the new derivatives. Their mode of action included an induction of apoptotic capsase-3 activity, while no contribution of unspecific cytotoxic effects was observed in LDH-release measurements. Kinase profiling of cancer relevant protein kinases identified the two 3-aryl-2-(thien-2-yl)acrylonitrile derivatives 1b and 1c as (multi-)kinase inhibitors with a preferential activity against the VEGFR-2 tyrosine kinase. Additional bioinformatic analysis of the VEGFR-2 binding modes by docking and molecular dynamics calculations supported the experimental findings and indicated that the hydroxy group of 1c might be crucial for its distinct inhibitory potency against VEGFR-2. Forthcoming studies will further unveil the underlying mode of action of the promising new derivatives as well as their suitability as an urgently needed novel approach in HCC treatment.


Assuntos
Acrilonitrila/química , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Tiofenos/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proliferação de Células , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Simulação de Acoplamento Molecular , Estrutura Molecular , Relação Estrutura-Atividade , Tiofenos/química
10.
Molecules ; 26(3)2021 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-33530327

RESUMO

While selective inhibition is one of the key assets for a small molecule drug, many diseases can only be tackled by simultaneous inhibition of several proteins. An example where achieving selectivity is especially challenging are ligands targeting human kinases. This difficulty arises from the high structural conservation of the kinase ATP binding sites, the area targeted by most inhibitors. We investigated the possibility to identify novel small molecule ligands with pre-defined binding profiles for a series of kinase targets and anti-targets by in silico docking. The candidate ligands originating from these calculations were assayed to determine their experimental binding profiles. Compared to previous studies, the acquired hit rates were low in this specific setup, which aimed at not only selecting multi-target kinase ligands, but also designing out binding to anti-targets. Specifically, only a single profiled substance could be verified as a sub-micromolar, dual-specific EGFR/ErbB2 ligand that indeed avoided its selected anti-target BRAF. We subsequently re-analyzed our target choice and in silico strategy based on these findings, with a particular emphasis on the hit rates that can be expected from a given target combination. To that end, we supplemented the structure-based docking calculations with bioinformatic considerations of binding pocket sequence and structure similarity as well as ligand-centric comparisons of kinases. Taken together, our results provide a multi-faceted picture of how pocket space can determine the success of docking in multi-target drug discovery efforts.


Assuntos
Simulação de Acoplamento Molecular/métodos , Proteínas Quinases/química , Proteínas Quinases/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Simulação por Computador , Descoberta de Drogas , Receptores ErbB/química , Receptores ErbB/metabolismo , Humanos , Ligantes , Modelos Moleculares , Conformação Molecular , Proteínas Proto-Oncogênicas B-raf/química , Proteínas Proto-Oncogênicas B-raf/metabolismo , Relação Estrutura-Atividade
11.
J Chem Inf Model ; 60(12): 6081-6094, 2020 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-33155465

RESUMO

Protein kinases play a crucial role in many cell signaling processes, making them one of the most important families of drug targets. In this context, fragment-based drug design strategies have been successfully applied to develop novel kinase inhibitors. These strategies usually follow a knowledge-driven approach to optimize a focused set of fragments to a potent kinase inhibitor. Alternatively, KinFragLib explores and extends the chemical space of kinase inhibitors using data-driven fragmentation and recombination. The method builds on available structural kinome data from the KLIFS database for over 2500 kinase DFG-in structures cocrystallized with noncovalent kinase ligands. The computational fragmentation method splits the ligands into fragments with respect to their 3D proximity to six predefined functionally relevant subpocket centers. The resulting fragment library consists of six subpocket pools with over 7000 fragments, available at https://github.com/volkamerlab/KinFragLib. KinFragLib offers two main applications: on the one hand, in-depth analyses of the chemical space of known kinase inhibitors, subpocket characteristics, and connections, and on the other hand, subpocket-informed recombination of fragments to generate potential novel inhibitors. The latter showed that recombining only a subset of 624 representative fragments generated 6.7 million molecules. This combinatorial library contains, besides some known kinase inhibitors, more than 99% novel chemical matter compared to ChEMBL and 63% molecules compliant with Lipinski's rule of five.


Assuntos
Inibidores de Proteínas Quinases , Proteínas Quinases , Desenho de Fármacos , Ligantes , Inibidores de Proteínas Quinases/farmacologia , Recombinação Genética
12.
J Chem Inf Model ; 60(12): 6211-6227, 2020 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-33119284

RESUMO

Alchemical free-energy calculations are now widely used to drive or maintain potency in small-molecule lead optimization with a roughly 1 kcal/mol accuracy. Despite this, the potential to use free-energy calculations to drive optimization of compound selectivity among two similar targets has been relatively unexplored in published studies. In the most optimistic scenario, the similarity of binding sites might lead to a fortuitous cancellation of errors and allow selectivity to be predicted more accurately than affinity. Here, we assess the accuracy with which selectivity can be predicted in the context of small-molecule kinase inhibitors, considering the very similar binding sites of human kinases CDK2 and CDK9 as well as another series of ligands attempting to achieve selectivity between the more distantly related kinases CDK2 and ERK2. Using a Bayesian analysis approach, we separate systematic from statistical errors and quantify the correlation in systematic errors between selectivity targets. We find that, in the CDK2/CDK9 case, a high correlation in systematic errors suggests that free-energy calculations can have significant impact in aiding chemists in achieving selectivity, while in more distantly related kinases (CDK2/ERK2), the correlation in systematic error suggests that fortuitous cancellation may even occur between systems that are not as closely related. In both cases, the correlation in systematic error suggests that longer simulations are beneficial to properly balance statistical error with systematic error to take full advantage of the increase in apparent free-energy calculation accuracy in selectivity prediction.


Assuntos
Desenho de Fármacos , Simulação de Dinâmica Molecular , Teorema de Bayes , Sítios de Ligação , Humanos , Ligantes , Ligação Proteica , Termodinâmica
13.
J Comput Aided Mol Des ; 34(7): 731-746, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32297073

RESUMO

In drug development, late stage toxicity issues of a compound are the main cause of failure in clinical trials. In silico methods are therefore of high importance to guide the early design process to reduce time, costs and animal testing. Technical advances and the ever growing amount of available toxicity data enabled machine learning, especially neural networks, to impact the field of predictive toxicology. In this study, cytotoxicity prediction, one of the earliest handles in drug discovery, is investigated using a deep learning approach trained on a highly consistent in-house data set of over 34,000 compounds with a share of less than 5% of cytotoxic molecules. The model reached a balanced accuracy of over 70%, similar to previously reported studies using Random Forest. Albeit yielding good results, neural networks are often described as a black box lacking deeper mechanistic understanding of the underlying model. To overcome this absence of interpretability, a Deep Taylor Decomposition method is investigated to identify substructures that may be responsible for the cytotoxic effects, the so-called toxicophores. Furthermore, this study introduces cytotoxicity maps which provide a visual structural interpretation of the relevance of these substructures. Using this approach could be helpful in drug development to predict the potential toxicity of a compound as well as to generate new insights into the toxic mechanism. Moreover, it could also help to de-risk and optimize compounds.


Assuntos
Citotoxinas/química , Citotoxinas/toxicidade , Aprendizado Profundo , Descoberta de Drogas/métodos , Sobrevivência Celular/efeitos dos fármacos , Desenho Assistido por Computador , Desenho de Fármacos , Descoberta de Drogas/estatística & dados numéricos , Células HEK293 , Células Hep G2 , Humanos , Modelos Biológicos , Redes Neurais de Computação , Bibliotecas de Moléculas Pequenas , Software , Toxicologia/estatística & dados numéricos
14.
J Chem Inf Model ; 59(10): 4083-4086, 2019 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-31612715

RESUMO

Open-source workflows have become more and more an integral part of computer-aided drug design (CADD) projects since they allow reproducible and shareable research that can be easily transferred to other projects. Setting up, understanding, and applying such workflows involves either coding or using workflow managers that offer a graphical user interface. We previously reported the TeachOpenCADD teaching platform that provides interactive Jupyter Notebooks (talktorials) on central CADD topics using open-source data and Python packages. Here we present the conversion of these talktorials to KNIME workflows that allow users to explore our teaching material without any line of code. TeachOpenCADD KNIME workflows are freely available on the KNIME Hub: https://hub.knime.com/volkamerlab/space/TeachOpenCADD .


Assuntos
Desenho de Fármacos , Modelos Químicos , Software , Fluxo de Trabalho , Simulação por Computador
15.
J Chem Inf Model ; 59(5): 1728-1742, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-30817146

RESUMO

Target deconvolution is a vital initial step in preclinical drug development to determine research focus and strategy. In this respect, computational target prediction is used to identify the most probable targets of an orphan ligand or the most similar targets to a protein under investigation. Applications range from the fundamental analysis of the mode-of-action over polypharmacology or adverse effect predictions to drug repositioning. Here, we provide a review on published ligand- and target-based as well as hybrid approaches for computational target prediction, together with current limitations and future directions.


Assuntos
Desenho de Fármacos , Descoberta de Drogas/métodos , Animais , Reposicionamento de Medicamentos/métodos , Humanos , Ligantes , Aprendizado de Máquina , Polifarmacologia , Mapas de Interação de Proteínas/efeitos dos fármacos , Proteínas/metabolismo
16.
Nucleic Acids Res ; 45(W1): W337-W343, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28472372

RESUMO

With currently more than 126 000 publicly available structures and an increasing growth rate, the Protein Data Bank constitutes a rich data source for structure-driven research in fields like drug discovery, crop science and biotechnology in general. Typical workflows in these areas involve manifold computational tools for the analysis and prediction of molecular functions. Here, we present the ProteinsPlus web server that offers a unified easy-to-use interface to a broad range of tools for the early phase of structure-based molecular modeling. This includes solutions for commonly required pre-processing tasks like structure quality assessment (EDIA), hydrogen placement (Protoss) and the search for alternative conformations (SIENA). Beyond that, it also addresses frequent problems as the generation of 2D-interaction diagrams (PoseView), protein-protein interface classification (HyPPI) as well as automatic pocket detection and druggablity assessment (DoGSiteScorer). The unified ProteinsPlus interface covering all featured approaches provides various facilities for intuitive input and result visualization, case-specific parameterization and download options for further processing. Moreover, its generalized workflow allows the user a quick familiarization with the different tools. ProteinsPlus also stores the calculated results temporarily for future request and thus facilitates convenient result communication and re-access. The server is freely available at http://proteins.plus.


Assuntos
Conformação Proteica , Software , Sítios de Ligação , Hidrogênio/química , Internet , Ligantes , Modelos Moleculares , Mapeamento de Interação de Proteínas , Proteínas/química
17.
Int J Mol Sci ; 20(2)2019 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-30658435

RESUMO

New inhibitors of tubulin polymerization and/or histone deacetylase (HDAC) activity were synthesized by attaching alkyl tethered hydroxamic acid appendages of varying length to oxazole-bridged combretastatin A-4 analogous caps. While their antiproliferative and microtubule disrupting effect was most pronounced for derivatives with short spacers, HDAC inhibition was strongest for those with longer spacers. These findings were further supported by computational methods such as structure-based docking experiments exploring the target interactions of the derivatives with varying linkers. For instance, compounds featuring short four-atom spacers between cap and hydroxamic acid inhibited the growth of various cancer cell lines and human endothelial hybrid cells with IC50 values in the low nanomolar range. In line with their ability to inhibit the microtubule assembly, four- and five-atom spacered hydroxamic acids caused an accumulation of 518A2 melanoma cells in G2/M phase, whereas a compound featuring a six-atom spacer and performing best in HDAC inhibition, induced a G1 arrest in these cells. All these beneficial anticancer activities together with their selectivity for cancer cells over non-malignant cells, point out the great potential of these novel pleiotropic HDAC and tubulin inhibitors as drug candidates for cancer therapy.


Assuntos
Bibenzilas/química , Bibenzilas/farmacologia , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/química , Oxazóis/química , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Sítios de Ligação , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Imunofluorescência , Humanos , Ligação de Hidrogênio , Concentração Inibidora 50 , Microtúbulos/metabolismo , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Ligação Proteica , Relação Estrutura-Atividade , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
18.
J Chem Inf Model ; 58(8): 1469-1472, 2018 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-30058337

RESUMO

Many doctoral students and postdoctoral fellows face at some point in their career the decision between continuing in academia or pursuing a job in industry. Both career paths come with their advantages and disadvantages as well as associated clichés. Our scientific journeys have led us from an university Ph.D. degree to an industrial postdoctoral stay and back to a young faculty position in academia. In this perspective, we share our experiences while changing perspectives. We will discuss the insights we gained through the phase as industrial postdoctoral fellows, the motivation to return and take up a young faculty position in academia, and the freedom and the burden of starting out as a principal investigator (PI). We end with our thoughts on "quo vadis" computational chemistry.


Assuntos
Escolha da Profissão , Biologia Computacional , Descoberta de Drogas , Indústria Farmacêutica , Pesquisadores , Mobilidade Ocupacional , Química , Docentes , Humanos , Pesquisa , Local de Trabalho
19.
Molecules ; 23(8)2018 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-30082611

RESUMO

Pharmacophore models are an accurate and minimal tridimensional abstraction of intermolecular interactions between chemical structures, usually derived from a group of molecules or from a ligand-target complex. Only a limited amount of solutions exists to model comprehensive pharmacophores using the information of a particular target structure without knowledge of any binding ligand. In this work, an automated and customable tool for truly target-focused (T²F) pharmacophore modeling is introduced. Key molecular interaction fields of a macromolecular structure are calculated using the AutoGRID energy functions. The most relevant points are selected by a newly developed filtering cascade and clustered to pharmacophore features with a density-based algorithm. Using five different protein classes, the ability of this method to identify essential pharmacophore features was compared to structure-based pharmacophores derived from ligand-target interactions. This method represents an extremely valuable instrument for drug design in a situation of scarce ligand information available, but also in the case of underexplored therapeutic targets, as well as to investigate protein allosteric pockets and protein-protein interactions.


Assuntos
Modelos Teóricos , Algoritmos , Desenho de Fármacos , Estrutura Molecular
20.
BMC Bioinformatics ; 18(1): 16, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-28056780

RESUMO

BACKGROUND: Annotations of the phylogenetic tree of the human kinome is an intuitive way to visualize compound profiling data, structural features of kinases or functional relationships within this important class of proteins. The increasing volume and complexity of kinase-related data underlines the need for a tool that enables complex queries pertaining to kinase disease involvement and potential therapeutic uses of kinase inhibitors. RESULTS: Here, we present KinMap, a user-friendly online tool that facilitates the interactive navigation through kinase knowledge by linking biochemical, structural, and disease association data to the human kinome tree. To this end, preprocessed data from freely-available sources, such as ChEMBL, the Protein Data Bank, and the Center for Therapeutic Target Validation platform are integrated into KinMap and can easily be complemented by proprietary data. The value of KinMap will be exemplarily demonstrated for uncovering new therapeutic indications of known kinase inhibitors and for prioritizing kinases for drug development efforts. CONCLUSION: KinMap represents a new generation of kinome tree viewers which facilitates interactive exploration of the human kinome. KinMap enables generation of high-quality annotated images of the human kinome tree as well as exchange of kinome-related data in scientific communications. Furthermore, KinMap supports multiple input and output formats and recognizes alternative kinase names and links them to a unified naming scheme, which makes it a useful tool across different disciplines and applications. A web-service of KinMap is freely available at http://www.kinhub.org/kinmap/ .


Assuntos
Bases de Dados de Proteínas , Internet , Proteínas Quinases/química , Software , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Desenho de Fármacos , Humanos , Modelos Moleculares , Biologia Molecular , Anotação de Sequência Molecular , Filogenia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA