Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Med Chem ; 63(23): 14425-14447, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33140646

RESUMEN

This article summarizes the evolution of the screening deck at the Novartis Institutes for BioMedical Research (NIBR). Historically, the screening deck was an assembly of all available compounds. In 2015, we designed a first deck to facilitate access to diverse subsets with optimized properties. We allocated the compounds as plated subsets on a 2D grid with property based ranking in one dimension and increasing structural redundancy in the other. The learnings from the 2015 screening deck were applied to the design of a next generation in 2019. We found that using traditional leadlikeness criteria (mainly MW, clogP) reduces the hit rates of attractive chemical starting points in subset screening. Consequently, the 2019 deck relies on solubility and permeability to select preferred compounds. The 2019 design also uses NIBR's experimental assay data and inferred biological activity profiles in addition to structural diversity to define redundancy across the compound sets.


Asunto(s)
Bibliotecas de Moléculas Pequeñas/química , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Bibliotecas de Moléculas Pequeñas/farmacología
2.
Sci Rep ; 10(1): 9670, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32541899

RESUMEN

Multiplexed gene-signature-based phenotypic assays are increasingly used for the identification and profiling of small molecule-tool compounds and drugs. Here we introduce a method (provided as R-package) for the quantification of the dose-response potency of a gene-signature as EC50 and IC50 values. Two signaling pathways were used as models to validate our methods: beta-adrenergic agonistic activity on cAMP generation (dedicated dataset generated for this study) and EGFR inhibitory effect on cancer cell viability. In both cases, potencies derived from multi-gene expression data were highly correlated with orthogonal potencies derived from cAMP and cell growth readouts, and superior to potencies derived from single individual genes. Based on our results we propose gene-signature potencies as a novel valid alternative for the quantitative prioritization, optimization and development of novel drugs.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias/genética , Agonistas Adrenérgicos beta/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Humanos , Concentración 50 Inhibidora , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Fenotipo , Transducción de Señal/efectos de los fármacos , Células THP-1
3.
Cell Chem Biol ; 26(5): 765-777.e3, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-30956147

RESUMEN

Libraries of well-annotated small molecules have many uses in chemical genetics, drug discovery, and therapeutic repurposing. Multiple libraries are available, but few data-driven approaches exist to compare them and design new libraries. We describe an approach to scoring and creating libraries based on binding selectivity, target coverage, and induced cellular phenotypes as well as chemical structure, stage of clinical development, and user preference. The approach, available via the online tool http://www.smallmoleculesuite.org, assembles sets of compounds with the lowest possible off-target overlap. Analysis of six kinase inhibitor libraries using our approach reveals dramatic differences among them and led us to design a new LSP-OptimalKinase library that outperforms existing collections in target coverage and compact size. We also describe a mechanism of action library that optimally covers 1,852 targets in the liganded genome. Our tools facilitate creation, analysis, and updates of both private and public compound collections.


Asunto(s)
Quimioinformática/métodos , Bibliotecas de Moléculas Pequeñas/análisis , Interfaz Usuario-Computador , Descubrimiento de Drogas , Inhibidores de Proteínas Quinasas/análisis , Inhibidores de Proteínas Quinasas/química , Bibliotecas de Moléculas Pequeñas/química
4.
J Med Chem ; 61(22): 10155-10172, 2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30339381

RESUMEN

SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin subfamily A member 2 (SMARCA2), also known as Brahma homologue (BRM), is a Snf2-family DNA-dependent ATPase. BRM and its close homologue Brahma-related gene 1 (BRG1), also known as SMARCA4, are mutually exclusive ATPases of the large ATP-dependent SWI/SNF chromatin-remodeling complexes involved in transcriptional regulation of gene expression. No small molecules have been reported that modulate SWI/SNF chromatin-remodeling activity via inhibition of its ATPase activity, an important goal given the well-established dependence of BRG1-deficient cancers on BRM. Here, we describe allosteric dual BRM and BRG1 inhibitors that downregulate BRM-dependent gene expression and show antiproliferative activity in a BRG1-mutant-lung-tumor xenograft model upon oral administration. These compounds represent useful tools for understanding the functions of BRM in BRG1-loss-of-function settings and should enable probing the role of SWI/SNF functions more broadly in different cancer contexts and those of other diseases.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , ADN Helicasas/genética , Diseño de Fármacos , Mutación , Proteínas Nucleares/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Administración Oral , Animales , Antineoplásicos/química , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Modelos Moleculares , Conformación Proteica , Relación Estructura-Actividad , Factores de Transcripción/química , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Elife ; 62017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28786378

RESUMEN

The Food and Drug Administration Adverse Event Reporting System (FAERS) remains the primary source for post-marketing pharmacovigilance. The system is largely un-curated, unstandardized, and lacks a method for linking drugs to the chemical structures of their active ingredients, increasing noise and artefactual trends. To address these problems, we mapped drugs to their ingredients and used natural language processing to classify and correlate drug events. Our analysis exposed key idiosyncrasies in FAERS, for example reports of thalidomide causing a deadly ADR when used against myeloma, a likely result of the disease itself; multiplications of the same report, unjustifiably increasing its importance; correlation of reported ADRs with public events, regulatory announcements, and with publications. Comparing the pharmacological, pharmacokinetic, and clinical ADR profiles of methylphenidate, aripiprazole, and risperidone, and of kinase drugs targeting the VEGF receptor, demonstrates how underlying molecular mechanisms can emerge from ADR co-analysis. The precautions and methods we describe may enable investigators to avoid confounding chemistry-based associations and reporting biases in FAERS, and illustrate how comparative analysis of ADRs can reveal underlying mechanisms.


Asunto(s)
Sistemas de Registro de Reacción Adversa a Medicamentos , Fenómenos Farmacológicos , Vigilancia de Productos Comercializados , Humanos , Estados Unidos , United States Food and Drug Administration
6.
Nat Chem Biol ; 11(12): 958-66, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26479441

RESUMEN

High-throughput screening (HTS) is an integral part of early drug discovery. Herein, we focused on those small molecules in a screening collection that have never shown biological activity despite having been exhaustively tested in HTS assays. These compounds are referred to as 'dark chemical matter' (DCM). We quantified DCM, validated it in quality control experiments, described its physicochemical properties and mapped it into chemical space. Through analysis of prospective reporter-gene assay, gene expression and yeast chemogenomics experiments, we evaluated the potential of DCM to show biological activity in future screens. We demonstrated that, despite the apparent lack of activity, occasionally these compounds can result in potent hits with unique activity and clean safety profiles, which makes them valuable starting points for lead optimization efforts. Among the identified DCM hits was a new antifungal chemotype with strong activity against the pathogen Cryptococcus neoformans but little activity at targets relevant to human safety.


Asunto(s)
Antifúngicos/farmacología , Cryptococcus neoformans/efectos de los fármacos , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Antifúngicos/química , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Relación Estructura-Actividad
7.
J Chem Inf Model ; 55(5): 956-62, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25915687

RESUMEN

High Throughput Screening (HTS) is a common approach in life sciences to discover chemical matter that modulates a biological target or phenotype. However, low assay throughput, reagents cost, or a flowchart that can deal with only a limited number of hits may impair screening large numbers of compounds. In this case, a subset of compounds is assayed, and in silico models are utilized to aid in iterative screening design, usually to expand around the found hits and enrich subsequent rounds for relevant chemical matter. However, this may lead to an overly narrow focus, and the diversity of compounds sampled in subsequent iterations may suffer. Active learning has been recently successfully applied in drug discovery with the goal of sampling diverse chemical space to improve model performance. Here we introduce a robust and straightforward iterative screening protocol based on naïve Bayes models. Instead of following up on the compounds with the highest scores in the in silico model, we pursue compounds with very low but positive values. This includes unique chemotypes of weakly active compounds that enhance the applicability domain of the model and increase the cumulative hit rates. We show in a retrospective application to 81 Novartis assays that this protocol leads to consistently higher compound and scaffold hit rates compared to a standard expansion around hits or an active learning approach. We recommend using the weak reinforcement strategy introduced herein for iterative screening workflows.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Aprendizaje Automático , Algoritmos , Teorema de Bayes , Simulación por Computador
8.
Drug Discov Today ; 20(4): 422-34, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25463034

RESUMEN

Vast amounts of bioactivity data have been generated for small molecules across public and corporate domains. Biological signatures, either derived from systematic profiling efforts or from existing historical assay data, have been successfully employed for small molecule mechanism-of-action elucidation, drug repositioning, hit expansion and screening subset design. This article reviews different types of biological descriptors and applications, and we demonstrate how biological data can outlive the original purpose or project for which it was generated. By comparing 150 HTS campaigns run at Novartis over the past decade on the basis of their active and inactive chemical matter, we highlight the opportunities and challenges associated with cross-project learning in drug discovery.


Asunto(s)
Minería de Datos , Bases de Datos de Compuestos Químicos , Bases de Datos Farmacéuticas , Descubrimiento de Drogas/métodos , Preparaciones Farmacéuticas/química , Animales , Simulación por Computador , Minería de Datos/historia , Bases de Datos de Compuestos Químicos/historia , Bases de Datos Farmacéuticas/historia , Descubrimiento de Drogas/historia , Historia del Siglo XXI , Humanos , Modelos Moleculares , Estructura Molecular , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
9.
AAPS J ; 16(4): 847-59, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24871342

RESUMEN

Close structural relationships between approved drugs and bioactive compounds were systematically assessed using matched molecular pairs. For structural analogs of drugs, target information was assembled from ChEMBL and compared to drug targets reported in DrugBank. For many drugs, multiple analogs were identified that were active against different targets. Some of these additional targets were closely related to known drug targets while others were not. Surprising discrepancies between reported drug targets and targets of close structural analogs were often observed. On one hand, the results suggest that hypotheses concerning alternative drug targets can often be formulated on the basis of close structural relationships to bioactive compounds that are easily detectable. It is conceivable that such obvious structure-target relationships are frequently not considered (or might be overlooked) when compounds are developed with a focus on a primary target and a few related (or undesired) ones. On the other hand, our findings also raise questions concerning database content and drug repositioning efforts.


Asunto(s)
Bases de Datos Farmacéuticas/estadística & datos numéricos , Preparaciones Farmacéuticas/química , Bases de Datos Farmacéuticas/normas , Sistemas de Liberación de Medicamentos , Reposicionamiento de Medicamentos , Humanos , Reproducibilidad de los Resultados , Relación Estructura-Actividad
10.
ACS Chem Biol ; 9(7): 1622-31, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24802392

RESUMEN

Computational target prediction methods using chemical descriptors have been applied exhaustively in drug discovery to elucidate the mechanisms-of-action (MOAs) of small molecules. To predict truly novel and unexpected small molecule-target interactions, compounds must be compared by means other than their chemical structure alone. Here we investigated predictions made by a method, HTS fingerprints (HTSFPs), that matches patterns of activities in experimental screens. Over 1,400 drugs and 1,300 natural products (NPs) were screened in more than 200 diverse assays, creating encodable activity patterns. The comparison of these activity patterns to an MOA-annotated reference panel led to the prediction of 5,281 and 2,798 previously unknown targets for the NP and drug sets, respectively. Intriguingly, there was limited overlap among the targets predicted; the drugs were more biased toward membrane receptors and the NPs toward soluble enzymes, consistent with the idea that they represent unexplored pharmacologies. Importantly, HTSFPs inferred targets that were beyond the prediction capabilities of standard chemical descriptors, especially for NPs but also for the more explored drug set. Of 65 drug-target predictions that we tested in vitro, 48 (73.8%) were confirmed with AC50 values ranging from 38 nM to 29 µM. Among these interactions was the inhibition of cyclooxygenases 1 and 2 by the HIV protease inhibitor Tipranavir. These newly discovered targets that are phylogenetically and phylochemically distant to the primary target provide an explanation for spontaneous bleeding events observed for patients treated with this drug, a physiological effect that was previously difficult to reconcile with the drug's known MOA.


Asunto(s)
Productos Biológicos/química , Productos Biológicos/farmacología , Descubrimiento de Drogas/métodos , Preparaciones Farmacéuticas/química , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Bases de Datos Farmacéuticas , Humanos , Modelos Moleculares , Terapia Molecular Dirigida , Farmacología
11.
J Med Chem ; 56(21): 8879-91, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24117015

RESUMEN

We introduce a novel strategy to sample bioactive chemical space, which follows-up on hits from fragment campaigns without the need for a crystal structure. Our results strongly suggest that screening a few hundred or thousand fragments can substantially improve the selection of small-molecule screening subsets. By combining fragment-based screening with virtual fragment linking and HTS fingerprints, we have developed an effective strategy not only to expand from low-affinity hits to potent compounds but also to hop in chemical space to substantially novel chemotypes. In benchmark calculations, our approach accessed subsets of compounds that were substantially enriched in chemically diverse hit compounds for various activity classes. Overall, half of the hits in the screening collection were found by screening only 10% of the library. Furthermore, a prospective application led to the discovery of two structurally novel histone deacetylase 4 inhibitors.


Asunto(s)
Inhibidores Enzimáticos/química , Bibliotecas de Moléculas Pequeñas/química , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Enzimas/metabolismo , Ensayos Analíticos de Alto Rendimiento , Modelos Moleculares , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
12.
Drug Discov Today ; 18(13-14): 674-80, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23454345

RESUMEN

How is the 'diversity' of a compound set defined and how is the most appropriate compound subset identified for assay when screening the entire HTS deck is not an option? A common approach has so far been to cover as much of the chemical space as possible by screening a chemically diverse set of compounds. We show that, rather than chemical diversity, the biologic diversity of a compound library is an essential requirement for hit identification. We describe a simple and efficient approach for the design of a HTS library based on compound-target diversity. Biodiverse compound subsets outperform chemically diverse libraries regarding hit rate and the total number of unique chemical scaffolds present among hits. Specifically, by screening ~19% of a HTS collection, we expect to discover ~50-80% of all desired bioactive compounds.


Asunto(s)
Minería de Datos , Bases de Datos de Compuestos Químicos , Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento , Preparaciones Farmacéuticas/química , Farmacología , Bibliotecas de Moléculas Pequeñas , Algoritmos , Animales , Humanos , Estructura Molecular , Terapia Molecular Dirigida , Estudios Retrospectivos , Relación Estructura-Actividad
13.
J Chem Inf Model ; 53(3): 692-703, 2013 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-23461561

RESUMEN

Virtual screening using bioactivity profiles has become an integral part of currently applied hit finding methods in pharmaceutical industry. However, a significant drawback of this approach is that it is only applicable to compounds that have been biologically tested in the past and have sufficient activity annotations for meaningful profile comparisons. Although bioactivity data generated in pharmaceutical institutions are growing on an unprecedented scale, the number of biologically annotated compounds still covers only a minuscule fraction of chemical space. For a newly synthesized compound or an isolated natural product to be biologically characterized across multiple assays, it may take a considerable amount of time. Consequently, this chemical matter will not be included in virtual screening campaigns based on bioactivity profiles. To overcome this problem, we herein introduce bioturbo similarity searching that uses chemical similarity to map molecules without biological annotations into bioactivity space and then searches for biologically similar compounds in this reference system. In benchmark calculations on primary screening data, we demonstrate that our approach generally achieves higher hit rates and identifies structurally more diverse compounds than approaches using chemical information only. Furthermore, our method is able to discover hits with novel modes of inhibition that traditional 2D and 3D similarity approaches are unlikely to discover. Test calculations on a set of natural products reveal the practical utility of the approach for identifying novel and synthetically more accessible chemical matter.


Asunto(s)
Algoritmos , Ensayos Analíticos de Alto Rendimiento/métodos , Benchmarking , Minería de Datos , Modelos Químicos , Modelos Moleculares , Conformación Molecular , Mapeo Peptídico , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad , Interfaz Usuario-Computador
14.
Proc Natl Acad Sci U S A ; 109(28): 11178-83, 2012 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-22711801

RESUMEN

Notwithstanding their key roles in therapy and as biological probes, 7% of approved drugs are purported to have no known primary target, and up to 18% lack a well-defined mechanism of action. Using a chemoinformatics approach, we sought to "de-orphanize" drugs that lack primary targets. Surprisingly, targets could be easily predicted for many: Whereas these targets were not known to us nor to the common databases, most could be confirmed by literature search, leaving only 13 Food and Drug Administration-approved drugs with unknown targets; the number of drugs without molecular targets likely is far fewer than reported. The number of worldwide drugs without reasonable molecular targets similarly dropped, from 352 (25%) to 44 (4%). Nevertheless, there remained at least seven drugs for which reasonable mechanism-of-action targets were unknown but could be predicted, including the antitussives clemastine, cloperastine, and nepinalone; the antiemetic benzquinamide; the muscle relaxant cyclobenzaprine; the analgesic nefopam; and the immunomodulator lobenzarit. For each, predicted targets were confirmed experimentally, with affinities within their physiological concentration ranges. Turning this question on its head, we next asked which drugs were specific enough to act as chemical probes. Over 100 drugs met the standard criteria for probes, and 40 did so by more stringent criteria. A chemical information approach to drug-target association can guide therapeutic development and reveal applications to probe biology, a focus of much current interest.


Asunto(s)
Biología Computacional/métodos , Tecnología Farmacéutica/métodos , Bases de Datos Factuales , Relación Dosis-Respuesta a Droga , Aprobación de Drogas , Sistemas de Liberación de Medicamentos , Humanos , Cinética , Ligandos , Sondas Moleculares/química , Preparaciones Farmacéuticas/química , Programas Informáticos , Estados Unidos , United States Food and Drug Administration , ortoaminobenzoatos/química
15.
Nature ; 486(7403): 361-7, 2012 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-22722194

RESUMEN

Discovering the unintended 'off-targets' that predict adverse drug reactions is daunting by empirical methods alone. Drugs can act on several protein targets, some of which can be unrelated by conventional molecular metrics, and hundreds of proteins have been implicated in side effects. Here we use a computational strategy to predict the activity of 656 marketed drugs on 73 unintended 'side-effect' targets. Approximately half of the predictions were confirmed, either from proprietary databases unknown to the method or by new experimental assays. Affinities for these new off-targets ranged from 1 nM to 30 µM. To explore relevance, we developed an association metric to prioritize those new off-targets that explained side effects better than any known target of a given drug, creating a drug-target-adverse drug reaction network. Among these new associations was the prediction that the abdominal pain side effect of the synthetic oestrogen chlorotrianisene was mediated through its newly discovered inhibition of the enzyme cyclooxygenase-1. The clinical relevance of this inhibition was borne out in whole human blood platelet aggregation assays. This approach may have wide application to de-risking toxicological liabilities in drug discovery.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Pruebas de Toxicidad/métodos , Plaquetas/efectos de los fármacos , Clorotrianiseno/efectos adversos , Clorotrianiseno/química , Clorotrianiseno/farmacología , Ciclooxigenasa 1/metabolismo , Inhibidores de la Ciclooxigenasa/efectos adversos , Inhibidores de la Ciclooxigenasa/farmacología , Bases de Datos Factuales , Estrógenos no Esteroides/efectos adversos , Estrógenos no Esteroides/farmacología , Predicción , Humanos , Modelos Biológicos , Terapia Molecular Dirigida/efectos adversos , Agregación Plaquetaria/efectos de los fármacos , Reproducibilidad de los Resultados , Especificidad por Sustrato
16.
ACS Chem Biol ; 7(8): 1399-409, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22594495

RESUMEN

Since the advent of high-throughput screening (HTS), there has been an urgent need for methods that facilitate the interrogation of large-scale chemical biology data to build a mode of action (MoA) hypothesis. This can be done either prior to the HTS by subset design of compounds with known MoA or post HTS by data annotation and mining. To enable this process, we developed a tool that compares compounds solely on the basis of their bioactivity: the chemical biological descriptor "high-throughput screening fingerprint" (HTS-FP). In the current embodiment, data are aggregated from 195 biochemical and cell-based assays developed at Novartis and can be used to identify bioactivity relationships among the in-house collection comprising ~1.5 million compounds. We demonstrate the value of the HTS-FP for virtual screening and in particular scaffold hopping. HTS-FP outperforms state of the art methods in several aspects, retrieving bioactive compounds with remarkable chemical dissimilarity to a probe structure. We also apply HTS-FP for the design of screening subsets in HTS. Using retrospective data, we show that a biodiverse selection of plates performs significantly better than a chemically diverse selection of plates, both in terms of number of hits and diversity of chemotypes retrieved. This is also true in the case of hit expansion predictions using HTS-FP similarity. Sets of compounds clustered with HTS-FP are biologically meaningful, in the sense that these clusters enrich for genes and gene ontology (GO) terms, showing that compounds that are bioactively similar also tend to target proteins that operate together in the cell. HTS-FP are valuable not only because of their predictive power but mainly because they relate compounds solely on the basis of bioactivity, harnessing the accumulated knowledge of a high-throughput screening facility toward the understanding of how compounds interact with the proteome.


Asunto(s)
Química Farmacéutica/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Animales , Bioquímica/métodos , Análisis por Conglomerados , Biología Computacional/métodos , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Ligandos , Modelos Químicos , Modelos Moleculares , Conformación Molecular , Relación Estructura-Actividad Cuantitativa
17.
Bioorg Med Chem ; 20(18): 5416-27, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22405595

RESUMEN

The increasing amount of chemogenomics data, that is, activity measurements of many compounds across a variety of biological targets, allows for better understanding of pharmacology in a broad biological context. Rather than assessing activity at individual biological targets, today understanding of compound interaction with complex biological systems and molecular pathways is often sought in phenotypic screens. This perspective poses novel challenges to structure-activity relationship (SAR) assessment. Today, the bottleneck of drug discovery lies in the understanding of SAR of rich datasets that go beyond single targets in the context of biological pathways, potential off-targets, and complex selectivity profiles. To aid in the understanding and interpretation of such complex SAR, we introduce Chemotography (chemotype chromatography), which encodes chemical space using a color spectrum by combining clustering and multidimensional scaling. Rich biological data in our approach were visualized using spatial dimensions traditionally reserved for chemical space. This allowed us to analyze SAR in the context of target hierarchies and phylogenetic trees, two-target activity scatter plots, and biological pathways. Chemotography, in combination with the Kyoto Encyclopedia of Genes and Genomes (KEGG), also allowed us to extract pathway-relevant SAR from the ChEMBL database. We identified chemotypes showing polypharmacology and selectivity-conferring scaffolds, even in cases where individual compounds have not been tested against all relevant targets. In addition, we analyzed SAR in ChEMBL across the entire Kinome, going beyond individual compounds. Our method combines the strengths of chemical space visualization for SAR analysis and graphical representation of complex biological data. Chemotography is a new paradigm for chemogenomic data visualization and its versatile applications presented here may allow for improved assessment of SAR in biological context, such as phenotypic assay hit lists.


Asunto(s)
Descubrimiento de Drogas , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Cromatografía , Análisis por Conglomerados , Bases de Datos Farmacéuticas , Estructura Molecular , Relación Estructura-Actividad
18.
Expert Opin Drug Metab Toxicol ; 7(12): 1497-511, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22050465

RESUMEN

INTRODUCTION: The goal of early predictive safety assessment (PSA) is to keep compounds with detectable liabilities from progressing further in the pipeline. Such compounds jeopardize the core of pharmaceutical research and development and limit the timely delivery of innovative therapeutics to the patient. Computational methods are increasingly used to help understand observed data, generate new testable hypotheses of relevance to safety pharmacology, and supplement and replace costly and time-consuming experimental procedures. AREAS COVERED: The authors survey methods operating on different scales of both physical extension and complexity. After discussing methods used to predict liabilities associated with structures of individual compounds, the article reviews the use of adverse event data and safety profiling panels. Finally, the authors examine the complexities of toxicology data from animal experiments and how these data can be mined. EXPERT OPINION: A significant obstacle for data-driven safety assessment is the absence of integrated data sets due to a lack of sharing of data and of using standard ontologies for data relevant to safety assessment. Informed decisions to derive focused sets of compounds can help to avoid compound liabilities in screening campaigns, and improved hit assessment of such campaigns can benefit the early termination of undesirable compounds.


Asunto(s)
Biología Computacional/métodos , Evaluación Preclínica de Medicamentos/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Preparaciones Farmacéuticas/metabolismo , Animales , Fenómenos Químicos , Simulación por Computador , Determinación de Punto Final , Humanos
19.
J Chem Inf Model ; 51(12): 3158-68, 2011 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-22098146

RESUMEN

From a medicinal chemistry point of view, one of the primary goals of high throughput screening (HTS) hit list assessment is the identification of chemotypes with an informative structure-activity relationship (SAR). Such chemotypes may enable optimization of the primary potency, as well as selectivity and phamacokinetic properties. A common way to prioritize them is molecular clustering of the hits. Typical clustering techniques, however, rely on a general notion of chemical similarity or standard rules of scaffold decomposition and are thus insensitive to molecular features that are enriched in biologically active compounds. This hinders SAR analysis, because compounds sharing the same pharmacophore might not end up in the same cluster and thus are not directly compared to each other by the medicinal chemist. Similarly, common chemotypes that are not related to activity may contaminate clusters, distracting from important chemical motifs. We combined molecular similarity and Bayesian models and introduce (I) a robust, activity-aware clustering approach and (II) a feature mapping method for the elucidation of distinct SAR determinants in polypharmacologic compounds. We evaluated the method on 462 dose-response assays from the Pubchem Bioassay repository. Activity-aware clustering grouped compounds sharing molecular cores that were specific for the target or pathway at hand, rather than grouping inactive scaffolds commonly found in compound series. Many of these core structures we also found in literature that discussed SARs of the respective targets. A numerical comparison of cores allowed for identification of the structural prerequisites for polypharmacology, i.e., distinct bioactive regions within a single compound, and pointed toward selectivity-conferring medchem strategies. The method presented here is generally applicable to any type of activity data and may help bridge the gap between hit list assessment and designing a medchem strategy.


Asunto(s)
Diseño de Fármacos , Ensayos Analíticos de Alto Rendimiento , Preparaciones Farmacéuticas/química , Relación Estructura-Actividad , Teorema de Bayes , Análisis por Conglomerados , Ensayos Analíticos de Alto Rendimiento/métodos , Farmacología
20.
Methods Mol Biol ; 672: 503-15, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20838982

RESUMEN

For chemical genetics and chemical biology, an important task is the identification of small molecules that are selective against individual targets and can be used as molecular probes for specific biological functions. To aid in the development of computational methods for selectivity analysis, molecular benchmark systems have been developed that capture compound selectivity data for pairs of targets. These molecular test systems are utilized for "selectivity searching" and the analysis of structure-selectivity relationships. Going beyond binary selectivity sets focusing on target pairs, a methodological framework, Molecular Formal Concept Analysis (MolFCA), is described for the definition and systematic mining of compound selectivity profiles.


Asunto(s)
Biología Computacional/métodos , Metodologías Computacionales , Diseño de Software , Benchmarking , Minería de Datos/métodos , Sondas Moleculares/química , Sondas Moleculares/genética , Estructura Molecular , Relación Estructura-Actividad , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...