Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 20812, 2024 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-39242880

RESUMEN

With the exponential progress in the field of cheminformatics, the conventional modeling approaches have so far been to employ supervised and unsupervised machine learning (ML) and deep learning models, utilizing the standard molecular descriptors, which represent the structural, physicochemical, and electronic properties of a particular compound. Deviating from the conventional approach, in this investigation, we have employed the classification Read-Across Structure-Activity Relationship (c-RASAR), which involves the amalgamation of the concepts of classification-based quantitative structure-activity relationship (QSAR) and Read-Across to incorporate Read-Across-derived similarity and error-based descriptors into a statistical and machine learning modeling framework. ML models developed from these RASAR descriptors use similarity-based information from the close source neighbors of a particular query compound. We have employed different classification modeling algorithms on the selected QSAR and RASAR descriptors to develop predictive models for efficient prediction of query compounds' hepatotoxicity. The predictivity of each of these models was evaluated on a large number of test set compounds. The best-performing model was also used to screen a true external data set. The concepts of explainable AI (XAI) coupled with Read-Across were used to interpret the contributions of the RASAR descriptors in the best c-RASAR model and to explain the chemical diversity in the dataset. The application of various unsupervised dimensionality reduction techniques like t-SNE and UMAP and the supervised ARKA framework showed the usefulness of the RASAR descriptors over the selected QSAR descriptors in their ability to group similar compounds, enhancing the modelability of the dataset and efficiently identifying activity cliffs. Furthermore, the activity cliffs were also identified from Read-Across by observing the nature of compounds constituting the nearest neighbors for a particular query compound. On comparing our simple linear c-RASAR model with the previously reported models developed using the same dataset derived from the US FDA Orange Book ( https://www.accessdata.fda.gov/scripts/cder/ob/index.cfm ), it was observed that our model is simple, reproducible, transferable, and highly predictive. The performance of the LDA c-RASAR model on the true external set supersedes that of the previously reported work. Therefore, the present simple LDA c-RASAR model can efficiently be used to predict the hepatotoxicity of query chemicals.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Relación Estructura-Actividad Cuantitativa , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Algoritmos , Aprendizaje Automático , Humanos , Quimioinformática/métodos
2.
J Cheminform ; 15(1): 47, 2023 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-37069675

RESUMEN

INTRODUCTION AND METHODOLOGY: Pairs of similar compounds that only differ by a small structural modification but exhibit a large difference in their binding affinity for a given target are known as activity cliffs (ACs). It has been hypothesised that QSAR models struggle to predict ACs and that ACs thus form a major source of prediction error. However, the AC-prediction power of modern QSAR methods and its quantitative relationship to general QSAR-prediction performance is still underexplored. We systematically construct nine distinct QSAR models by combining three molecular representation methods (extended-connectivity fingerprints, physicochemical-descriptor vectors and graph isomorphism networks) with three regression techniques (random forests, k-nearest neighbours and multilayer perceptrons); we then use each resulting model to classify pairs of similar compounds as ACs or non-ACs and to predict the activities of individual molecules in three case studies: dopamine receptor D2, factor Xa, and SARS-CoV-2 main protease. RESULTS AND CONCLUSIONS: Our results provide strong support for the hypothesis that indeed QSAR models frequently fail to predict ACs. We observe low AC-sensitivity amongst the evaluated models when the activities of both compounds are unknown, but a substantial increase in AC-sensitivity when the actual activity of one of the compounds is given. Graph isomorphism features are found to be competitive with or superior to classical molecular representations for AC-classification and can thus be employed as baseline AC-prediction models or simple compound-optimisation tools. For general QSAR-prediction, however, extended-connectivity fingerprints still consistently deliver the best performance amongs the tested input representations. A potential future pathway to improve QSAR-modelling performance might be the development of techniques to increase AC-sensitivity.

3.
Sci Total Environ ; 873: 162263, 2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-36801331

RESUMEN

Androgen mimicking environmental chemicals can bind to Androgen receptor (AR) and can cause severe effects on the reproductive health of males. Predicting such endocrine disrupting chemicals (EDCs) in the human exposome is vital for improving current chemical regulations. To this end, QSAR models have been developed to predict androgen binders. However, a continuous structure-activity relationship (SAR) wherein chemicals with similar structure have similar activity does not always hold. Activity landscape analysis can help map the structure-activity landscape and identify unique features such as activity cliffs. Here we performed a systematic investigation of the chemical diversity along with the global and local structure-activity landscape of a curated list of 144 AR binding chemicals. Specifically, we clustered the AR binding chemicals and visualized the associated chemical space. Thereafter, consensus diversity plot was used to assess the global diversity of the chemical space. Subsequently, the structure-activity landscape was investigated using SAS maps which capture the activity difference and structural similarity among the AR binders. This analysis led to a subset of 41 AR binding chemicals forming 86 activity cliffs, of which 14 are activity cliff generators. Additionally, SALI scores were computed for all pairs of AR binding chemicals and the SALI heatmap was also used to evaluate the activity cliffs identified using SAS map. Finally, we provide a classification of the 86 activity cliffs into six categories using structural information of chemicals at different levels. Overall, this investigation reveals the heterogeneous nature of the structure-activity landscape of AR binding chemicals and provides insights which will be crucial in preventing false prediction of chemicals as androgen binders and developing predictive computational toxicity models in the future.


Asunto(s)
Andrógenos , Receptores Androgénicos , Humanos , Receptores Androgénicos/metabolismo , Relación Estructura-Actividad , Relación Estructura-Actividad Cuantitativa
4.
Molecules ; 28(2)2023 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-36677547

RESUMEN

Currently, G protein-coupled receptors (GPCRs) constitute a significant group of membrane-bound receptors representing more than 30% of therapeutic targets. Fluorine is commonly used in designing highly active biological compounds, as evidenced by the steadily increasing number of drugs by the Food and Drug Administration (FDA). Herein, we identified and analyzed 898 target-based F-containing isomeric analog sets for SAR analysis in the ChEMBL database-FiSAR sets active against 33 different aminergic GPCRs comprising a total of 2163 fluorinated (1201 unique) compounds. We found 30 FiSAR sets contain activity cliffs (ACs), defined as pairs of structurally similar compounds showing significant differences in affinity (≥50-fold change), where the change of fluorine position may lead up to a 1300-fold change in potency. The analysis of matched molecular pair (MMP) networks indicated that the fluorination of aromatic rings showed no clear trend toward a positive or negative effect on affinity. Additionally, we propose an in silico workflow (including induced-fit docking, molecular dynamics, quantum polarized ligand docking, and binding free energy calculations based on the Generalized-Born Surface-Area (GBSA) model) to score the fluorine positions in the molecule.


Asunto(s)
Flúor , Simulación de Dinámica Molecular , Flúor/química , Unión Proteica , Receptores Acoplados a Proteínas G/química , Isomerismo , Ligandos , Simulación del Acoplamiento Molecular
5.
Mol Inform ; 41(11): e2200049, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35973966

RESUMEN

Activity cliffs (ACs) are defined as pairs of structurally similar compounds with large difference in their potencies against certain biotarget. We recently proposed that potent AC members induce significant entropically-driven conformational modifications of the target that unveil additional binding interactions, while their weakly-potent counterparts are enthalpically-driven binders with little influence on the protein target. We herein propose to extract pharmacophores for ACs-infested target(s) from molecular dynamics (MD) frames of purely "enthalpic" potent binder(s) complexed within the particular target. Genetic function algorithm/machine learning (GFA/ML) can then be employed to search for the best possible combination of MD pharmacophore(s) capable of explaining bioactivity variations within a list of inhibitors. We compared the performance of this approach with established ligand-based and structure-based methods. Kinase inserts domain receptor (KDR) was used as a case study. KDR plays a crucial role in angiogenic signalling and its inhibitors have been approved in cancer treatment. Interestingly, GFA/ML selected, MD-based, pharmacophores were of comparable performances to ligand-based and structure-based pharmacophores. The resulting pharmacophores and QSAR models were used to capture hits from the national cancer institute list of compounds. The most active hit showed anti-KDR IC50 of 2.76 µM.


Asunto(s)
Simulación de Dinámica Molecular , Relación Estructura-Actividad Cuantitativa , Ligandos
6.
J Comput Aided Mol Des ; 36(1): 39-62, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35059939

RESUMEN

Activity cliffs (ACs) are defined as closely analogous compounds of significant affinity discrepancies against certain biotarget. In this paper we propose to use AC pair(s) for extracting valid binding pharmacophores through exposing corresponding protein complexes to stochastic deformation/relaxation followed by applying genetic algorithm/machine learning (GA-ML) for selecting optimal pharmacophore(s) that best classify a long list of inhibitors. We compared the performances of ligand-based and structure-based pharmacophores with counterparts generated by this newly introduced technique. Sphingosine kinase 1 (SPHK-1) was used as case study. SPHK-1 is a lipid kinase that plays pivotal role in the regulation of a variety of biological processes including, cell growth, apoptosis, and inflammation. The new approach proved to yield pharmacophore and ML models of comparable accuracies to established ligand-based and structure-based pharmacophores. The resulting pharmacophores and ML models were used to capture hits from the national cancer institute list of compounds and predict their bioactivity categories. Two hits of novel chemotypes showed selective and low micromolar inhibitory IC50 values against SPHK-1.


Asunto(s)
Fosfotransferasas (Aceptor de Grupo Alcohol) , Relación Estructura-Actividad Cuantitativa , Ligandos , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología
7.
Chem Biol Drug Des ; 99(2): 308-319, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34806310

RESUMEN

Very small chemical changes in active compounds causing large potency effects are of particular interest in medicinal chemistry and drug design. We have systematically searched active compounds with available high-confidence activity data for pairs of structural analogs with dual-atom replacements and additional analogs with corresponding single-atom replacements. From ~287,000 unique qualifying compounds with activity against nearly 1900 unique targets, ~3500 target-based analog pairs with dual-atom replacements were identified. These included 852 pairs with significant differences in compound potency, representing a set of previously unobserved activity cliffs. Comparing these pairs with corresponding single-atom replacement analogs, which were frequently identified, made it possible to systematically analyze how potency changes propagated from single- to dual-atom replacements. The analysis uncovered different potency effects and revealed that individual atom replacements were often decisive for activity cliff formation. For a limited number of activity cliffs, X-ray structures of targets in complex with cliff compounds were available, which aided in rationalizing potency alterations among analogs with single- or dual-atom replacements. The analog pairs identified herein provide a rich resource of structure-activity relationship information and attractive test cases for calibrating computational methods.


Asunto(s)
Cristalografía por Rayos X/métodos , Diseño de Fármacos , Ensayos Analíticos de Alto Rendimiento , Estructura Molecular , Relación Estructura-Actividad
8.
Biomolecules ; 11(11)2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34827645

RESUMEN

Currently, G protein-coupled receptors are the targets with the highest number of drugs in many therapeutic areas. Fluorination has become a common strategy in designing highly active biological compounds, as evidenced by the steadily increasing number of newly approved fluorine-containing drugs. Herein, we identified in the ChEMBL database and analysed 1554 target-based FSAR sets (non-fluorinated compounds and their fluorinated analogues) comprising 966 unique non-fluorinated and 2457 unique fluorinated compounds active against 33 different aminergic GPCRs. Although a relatively small number of activity cliffs (defined as a pair of structurally similar compounds showing significant differences of activity -ΔpPot > 1.7) was found in FSAR sets, it is clear that appropriately introduced fluorine can increase ligand potency more than 50-fold. The analysis of matched molecular pairs (MMPs) networks indicated that the fluorination of the aromatic ring showed no clear trend towards a positive or negative effect on affinity; however, a favourable site for a positive potency effect of fluorination was the ortho position. Fluorination of aliphatic fragments more often led to a decrease in biological activity. The results may constitute the rules of thumb for fluorination of aminergic receptor ligands and provide insights into the role of fluorine substitutions in medicinal chemistry.


Asunto(s)
Receptores Acoplados a Proteínas G , Halogenación , Unión Proteica
9.
Environ Sci Technol ; 55(10): 6857-6866, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33914508

RESUMEN

Chemicals may cause adverse effects on human health through binding to peroxisome proliferator-activated receptor γ (PPARγ). Hence, binding affinity is useful for evaluating chemicals with potential endocrine-disrupting effects. Quantitative structure-activity relationship (QSAR) regression models with defined applicability domains (ADs) are important to enable efficient screening of chemicals with PPARγ binding activity. However, lack of large data sets hindered the development of QSAR models. In this study, based on PPARγ binding affinity data sets curated from various sources, 30 QSAR models were developed using molecular fingerprints, two-dimensional descriptors, and five machine learning algorithms. Structure-activity landscapes (SALs) of the training compounds were described by network-like similarity graphs (NSGs). Based on the NSGs, local discontinuity scores were calculated and found to be positively correlated with the cross-validation absolute prediction errors of the models using the different training sets, descriptors, and algorithms. Moreover, innovative ADs were defined based on pairwise similarities between compounds and were found to outperform some conventional ADs. The curated data sets and developed regression models could be useful for evaluating PPARγ-involved adverse effects of chemicals. The SAL analysis and the innovative ADs could facilitate understanding of prediction results from QSAR models.


Asunto(s)
PPAR gamma , Relación Estructura-Actividad Cuantitativa , Algoritmos , Humanos , Aprendizaje Automático , Unión Proteica
10.
J Comput Aided Mol Des ; 35(12): 1157-1164, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33740200

RESUMEN

An activity cliff (AC) is formed by a pair of structurally similar compounds with a large difference in potency. Accordingly, ACs reveal structure-activity relationship (SAR) discontinuity and provide SAR information for compound optimization. Herein, we have investigated the question if ACs could be predicted from image data. Therefore, pairs of structural analogs were extracted from different compound activity classes that formed or did not form ACs. From these compound pairs, consistently formatted images were generated. Image sets were used to train and test convolutional neural network (CNN) models to systematically distinguish between ACs and non-ACs. The CNN models were found to predict ACs with overall high accuracy, as assessed using alternative performance measures, hence establishing proof-of-principle. Moreover, gradient weights from convolutional layers were mapped to test compounds and identified characteristic structural features that contributed to successful predictions. Weight-based feature visualization revealed the ability of CNN models to learn chemistry from images at a high level of resolution and aided in the interpretation of model decisions with intrinsic black box character.


Asunto(s)
Diseño de Fármacos , Redes Neurales de la Computación , Relación Estructura-Actividad
11.
Molecules ; 26(3)2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33499015

RESUMEN

Tyrosinase is an enzyme that plays a crucial role in the melanogenesis of humans and the browning of food products. Thus, tyrosinase inhibitors that are useful to the cosmetic and food industries are required. In this study, we have used evolutionary chemical binding similarity (ECBS) to screen a virtual chemical database for human tyrosinase, which resulted in seven potential tyrosinase inhibitors confirmed through the tyrosinase inhibition assay. The tyrosinase inhibition percentage for three of the new actives was over 90% compared to 61.9% of kojic acid. From the structural analysis through pharmacophore modeling and molecular docking with the human tyrosinase model, the pi-pi interaction of tyrosinase inhibitors with conserved His367 and the polar interactions with Asn364, Glu345, and Glu203 were found to be essential for tyrosinase-ligand interactions. The pharmacophore features and the docking models showed high consistency, revealing the possible essential binding interactions of inhibitors to human tyrosinase. We have also presented the activity cliff analysis that successfully revealed the chemical features related to substantial activity changes found in the new tyrosinase inhibitors. The newly identified inhibitors and their structure-activity relationships presented here will help to identify or design new human tyrosinase inhibitors.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Monofenol Monooxigenasa/antagonistas & inhibidores , Dominio Catalítico/genética , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Humanos , Técnicas In Vitro , Ligandos , Simulación del Acoplamiento Molecular , Monofenol Monooxigenasa/química , Monofenol Monooxigenasa/genética , Pironas/química , Pironas/farmacología , Bibliotecas de Moléculas Pequeñas , Homología Estructural de Proteína , Relación Estructura-Actividad , Interfaz Usuario-Computador
12.
Data Brief ; 33: 106364, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33088875

RESUMEN

Activity cliffs (ACs) are defined as pairs of structurally similar or analogous active compounds with large potency differences [1]. As such, they provide important information for the exploration of structure-activity relationships (SARs) and chemical optimization. We have introduced a new category of ACs capturing minimal (single-atom) chemical modifications and identified more than 1500 of such ACs in compounds with activity against a variety of target proteins [2]. ACs with single-atom modifications (sam_ACs) include "atom-replacement ACs" (ar_ACs) that contain a single-atom replacement (N to C (N-C), O-C, N-O, or S-O) at a given position and "atom-walk ACs" (aw_ACs), in which two analogs are only distinguished by the position of a single heteroatom (non-carbon atom). For a number of sam_ACs, X-ray structures of complexes between AC targets and AC compounds were identified, which made it possible to explore the formation of sam_ACs on the basis of well-defined ligand-target interactions [2]. Our collection of sam_ACs including associated chemical and X-ray structure information, as described herein, is made freely available.

13.
Eur J Med Chem ; 207: 112846, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-32977219

RESUMEN

In medicinal chemistry, activity cliffs (ACs) are considered as sources of critical structure-activity relationship (SAR) information. ACs are capable of revealing such SAR information because they are formed by pairs or groups of structural analogs that are distinguished by small chemical modifications leading to large variations in compound potency. Such modifications can reveal critically important substitution sites in analog series. Small AC-encoded chemical changes enable the identification of SAR determinants. In this work, we have searched medicinal chemistry data for most "subtle" ACs in which participating compounds are only distinguished by single-atom modifications. These ACs can be directly associated with lead optimization strategies such as positional atom scanning (atom "walks") or heteroatom replacements in ring structures. More than 1500 of these ACs with activity against a variety of targets were identified. To further explore newly identified ACs, we searched for X-ray structures of ligand-target complexes containing participating AC compounds. For a subset of subtle ACs, X-ray structures of complexes made it possible to examine effects of single-atom changes in light of well-defined ligand-target interactions. Since ACs capturing minimal chemical changes are of particular interest for lead optimization and drug design, we make all newly identified ACs and associated structural information freely available as an open access deposition.


Asunto(s)
Química Farmacéutica , Diseño de Fármacos , Cristalografía por Rayos X , Minería de Datos , Bases de Datos Farmacéuticas , Ligandos
15.
J Comput Aided Mol Des ; 34(9): 943-952, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32500478

RESUMEN

Activity cliffs (ACs) consist of structurally similar compounds with a large difference in potency against their target. Accordingly, ACs introduce discontinuity in structure-activity relationships (SARs) and are a prime source of SAR information. In compound data sets, the vast majority of ACs are formed by differently sized groups of structurally similar compounds with large potency variations. As a consequence, many of these compounds participate in multiple ACs. This coordinated formation of ACs increases their SAR information content compared to ACs considered as individual compound pairs, but complicates AC analysis. In network representations, coordinated ACs give rise to clusters of varying size and topology, which can be interactively and computationally analyzed. While AC networks are indispensable tools to study coordinated ACs, they become difficult to navigate and interpret in the presence of clusters of increasing size and complex topologies. Herein, we introduce reduced network representations that transform AC networks into an easily interpretable format from which SAR information in the form of R-group tables can be readily obtained. The simplified network variant greatly improves the interpretability of large and complex AC networks and substantially supports SAR exploration.


Asunto(s)
Química Farmacéutica , Biología Computacional/métodos , Diseño de Fármacos , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Análisis por Conglomerados , Humanos , Estructura Molecular , Relación Estructura-Actividad
16.
Future Sci OA ; 6(5): FSO472, 2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32518687

RESUMEN

AIM: Extending the public knowledge base of activity cliffs (ACs) with new categories of ACs having special structural characteristics. METHODOLOGY: Dual-site ACs, isomer ACs and ACs with privileged substructures are described and their systematic identification is detailed. EXEMPLARY RESULTS & DATA: More than 7400 new ACs belonging to different categories with activity against more than 200 targets were identified and are made publicly available. LIMITATIONS & NEXT STEPS: For dual-site ACs, limited numbers of isomers are available as structural analogs for rationalizing contributions to AC formation. The search for such analogs will continue. In addition, the target distribution of ACs containing privileged substructures will be further analyzed.

17.
J Mol Graph Model ; 99: 107615, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32339898

RESUMEN

Janus kinase 1 (JAK1) is protein kinase involved in autoimmune diseases (AIDs). JAK1 inhibitors have shown promising results in treating AIDs. JAK1 inhibitors are known to exhibit regions of SAR discontinuity or activity cliffs (ACs). ACs represent fundamental challenge to successful QSAR/pharmacophore modeling because QSAR modeling rely on the basic premise that activity is a smooth continuous function of structure. We propose that ACs exist because active ACs members exhibit subtle, albeit critical, enthalpic features absent from their inactive twins. In this context we compared the performances of two computational modeling workflows in extracting valid pharmacophores from 151 diverse JAK1 inhibitors that include ACs: QSAR-guided pharmacophore selection versus docking-based comparative intermolecular contacts analysis (db-CICA). The two methods were judged based on the receiver operating characteristic (ROC) curves of their corresponding pharmacophore models and their abilities to distinguish active members among established JAK1 ACs. db-CICA modeling significantly outperformed ligand-based pharmacophore modeling. The resulting optimal db-CICA pharmacophore was used as virtual search query to scan the National Cancer Institute (NCI) database for novel JAK1 inhibitory leads. The most active hit showed IC50 of 1.04 µM. This study proposes the use of db-CICA modeling as means to extract valid pharmacophores from SAR data infested with ACs.


Asunto(s)
Inhibidores de Proteínas Quinasas , Relación Estructura-Actividad Cuantitativa , Janus Quinasa 1 , Ligandos , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/farmacología , Curva ROC
18.
J Comput Aided Mol Des ; 34(6): 659-669, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32060676

RESUMEN

In this work, we analyze the structure-activity relationships (SAR) of epigenetic inhibitors (lysine mimetics) against lysine methyltransferase (G9a or EHMT2) using a combined activity landscape, molecular docking and molecular dynamics approach. The study was based on a set of 251 G9a inhibitors with reported experimental activity. The activity landscape analysis rapidly led to the identification of activity cliffs, scaffolds hops and other active an inactive molecules with distinct SAR. Structure-based analysis of activity cliffs, scaffold hops and other selected active and inactive G9a inhibitors by means of docking followed by molecular dynamics simulations led to the identification of interactions with key residues involved in activity against G9a, for instance with ASP 1083, LEU 1086, ASP 1088, TYR 1154 and PHE 1158. The outcome of this work is expected to further advance the development of G9a inhibitors.


Asunto(s)
Inhibidores Enzimáticos/química , Antígenos de Histocompatibilidad/química , N-Metiltransferasa de Histona-Lisina/química , Relación Estructura-Actividad , Antígenos de Histocompatibilidad/ultraestructura , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/ultraestructura , Humanos , Lisina/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Conformación Proteica/efectos de los fármacos , Quinazolinas/química
19.
Mol Pharm ; 17(3): 979-989, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31978299

RESUMEN

The privileged substructure (PS) and activity cliff (AC) concepts are popular in pharmaceutical research. PSs have been empirically identified as preferred building blocks for target-class-directed generation of active compounds. Although some PSs are controversially viewed, they continue to receive much attention in drug discovery. ACs are formed by structurally similar active compounds with large potency differences and thus capture structure-activity relationship (SAR) discontinuity and reveal SAR determinants. So far, the PS and AC concepts have not been investigated in context. We have systematically explored ACs formed by compounds containing different PSs (PS-ACs). Such ACs were frequently identified in different series of compounds. PS-ACs were thoroughly characterized and compared to ACs formed by other compounds. The analysis revealed differences in AC formation between PSs. For example, individual PSs with an unusually high proportion of AC-forming compounds were identified. Furthermore, PS-AC network analysis identified clusters of ACs containing the same PS in different compound structure contexts with activity against different targets. From such PS-AC clusters, target-specific SAR information for PSs in different structural environments can be extracted.


Asunto(s)
Diseño de Fármacos , Descubrimiento de Drogas/métodos , Investigación Farmacéutica , Compuestos de Bifenilo/química , Proteínas Portadoras/química , Química Farmacéutica , Enzimas/química , Humanos , Indoles/química , Canales Iónicos/química , Estructura Molecular , Quinolinas/química , Receptores Citoplasmáticos y Nucleares/química , Relación Estructura-Actividad , Factores de Transcripción/química
20.
MethodsX ; 7: 100793, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31993342

RESUMEN

In medicinal chemistry and chemoinformatics, activity cliffs (ACs) are defined as pairs of structurally similar compounds that are active against the same target but have a large difference in potency. Accordingly, ACs are rich in structure-activity relationship (SAR) information, which rationalizes their relevance for medicinal chemistry. For identifying ACs, a compound similarity criterion and a potency difference criterion must be specified. So far a constant potency difference between AC partner compounds has mostly been set, e.g. 100-fold, irrespective of the specific activity (targets) of cliff-forming compounds. Herein, we introduce a computational methodology for AC identification and analysis that includes three novel components: •ACs are identified on the basis of variable target set-dependent potency difference criteria (a 'target set' represents a collection of compounds that are active against a given target protein).•ACs are extracted from computationally determined analog series (ASs) and consist of pairs of analogs with single or multiple substitution sites.•For multi-site ACs, a search for analogs with individual substitutions is performed to analyze their contributions to AC formation and determine if multi-site ACs can be represented by single-site ACs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA