Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.948
Filtrar
1.
Expert Rev Mol Med ; 26: e6, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38604802

RESUMO

Target deconvolution can help understand how compounds exert therapeutic effects and can accelerate drug discovery by helping optimise safety and efficacy, revealing mechanisms of action, anticipate off-target effects and identifying opportunities for therapeutic expansion. Chemoproteomics, a combination of chemical biology with mass spectrometry has transformed target deconvolution. This review discusses modification-free chemoproteomic approaches that leverage the change in protein thermodynamics induced by small molecule ligand binding. Unlike modification-based methods relying on enriching specific protein targets, these approaches offer proteome-wide evaluations, driven by advancements in mass spectrometry sensitivity, increasing proteome coverage and quantitation methods. Advances in methods based on denaturation/precipitation by thermal or chemical denaturation, or by protease degradation are evaluated, emphasising the evolving landscape of chemoproteomics and its potential impact on future drug-development strategies.


Assuntos
Descoberta de Drogas , Proteoma , Humanos , Proteoma/análise , Proteoma/química , Proteoma/metabolismo , Descoberta de Drogas/métodos , Espectrometria de Massas , Desenvolvimento de Medicamentos
2.
J Bioinform Comput Biol ; 22(1): 2450003, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38567386

RESUMO

In this paper, we propose a novel approach for predicting the activity/inactivity of molecules with the BRCA1 gene by combining pharmacophore modeling and deep learning techniques. Initially, we generated 3D pharmacophore fingerprints using a pharmacophore model, which captures the essential features and spatial arrangements critical for biological activity. These fingerprints served as informative representations of the molecular structures. Next, we employed deep learning algorithms to train a predictive model using the generated pharmacophore fingerprints. The deep learning model was designed to learn complex patterns and relationships between the pharmacophore features and the corresponding activity/inactivity labels of the molecules. By utilizing this integrated approach, we aimed to enhance the accuracy and efficiency of activity prediction. To validate the effectiveness of our approach, we conducted experiments using a dataset of known molecules with BRCA1 gene activity/inactivity from diverse sources. Our results demonstrated promising predictive performance, indicating the successful integration of pharmacophore modeling and deep learning. Furthermore, we utilized the trained model to predict the activity/inactivity of unknown molecules extracted from the ChEMBL database. The predictions obtained from the ChEMBL database were assessed and compared against experimentally determined values to evaluate the reliability and generalizability of our model. Overall, our proposed approach showcased significant potential in accurately predicting the activity/inactivity of molecules with the BRCA1 gene, thus enabling the identification of potential candidates for further investigation in drug discovery and development processes.


Assuntos
Aprendizado Profundo , Farmacóforo , Genes BRCA1 , Reprodutibilidade dos Testes , Descoberta de Drogas/métodos
3.
ACS Chem Biol ; 19(4): 938-952, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38565185

RESUMO

Phenotypic assays have become an established approach to drug discovery. Greater disease relevance is often achieved through cellular models with increased complexity and more detailed readouts, such as gene expression or advanced imaging. However, the intricate nature and cost of these assays impose limitations on their screening capacity, often restricting screens to well-characterized small compound sets such as chemogenomics libraries. Here, we outline a cheminformatics approach to identify a small set of compounds with likely novel mechanisms of action (MoAs), expanding the MoA search space for throughput limited phenotypic assays. Our approach is based on mining existing large-scale, phenotypic high-throughput screening (HTS) data. It enables the identification of chemotypes that exhibit selectivity across multiple cell-based assays, which are characterized by persistent and broad structure activity relationships (SAR). We validate the effectiveness of our approach in broad cellular profiling assays (Cell Painting, DRUG-seq, and Promotor Signature Profiling) and chemical proteomics experiments. These experiments revealed that the compounds behave similarly to known chemogenetic libraries, but with a notable bias toward novel protein targets. To foster collaboration and advance research in this area, we have curated a public set of such compounds based on the PubChem BioAssay dataset and made it available for use by the scientific community.


Assuntos
Descoberta de Drogas , Ensaios de Triagem em Larga Escala , Ensaios de Triagem em Larga Escala/métodos , Descoberta de Drogas/métodos
4.
Sci Rep ; 14(1): 7526, 2024 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-38565852

RESUMO

High throughput screening (HTS) is routinely used to identify bioactive small molecules. This requires physical compounds, which limits coverage of accessible chemical space. Computational approaches combined with vast on-demand chemical libraries can access far greater chemical space, provided that the predictive accuracy is sufficient to identify useful molecules. Through the largest and most diverse virtual HTS campaign reported to date, comprising 318 individual projects, we demonstrate that our AtomNet® convolutional neural network successfully finds novel hits across every major therapeutic area and protein class. We address historical limitations of computational screening by demonstrating success for target proteins without known binders, high-quality X-ray crystal structures, or manual cherry-picking of compounds. We show that the molecules selected by the AtomNet® model are novel drug-like scaffolds rather than minor modifications to known bioactive compounds. Our empirical results suggest that computational methods can substantially replace HTS as the first step of small-molecule drug discovery.


Assuntos
Descoberta de Drogas , Ensaios de Triagem em Larga Escala , Ensaios de Triagem em Larga Escala/métodos , Descoberta de Drogas/métodos , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/química
5.
J Med Chem ; 67(5): 3874-3884, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38426508

RESUMO

Fragment-based lead discovery has emerged as one of the most efficient screening strategies for finding hit molecules in drug discovery. Recently, a novel strategy based on a class of fragments characterized by an ultralow molecular weight (ULMW) has been proposed. These fragments bind to the target with a very low affinity, requiring reliable biophysical methods for detection. The most notable application of ULMW used a set of 81 fragments, named MiniFrags, and screened them by X-ray crystallography. We extended the utilization of this novel class of fragments to another gold standard technique for fragment-based screening: nuclear magnetic resonance (NMR). Here, we present a novel NMR protocol to detect and analyze such weak interactions in a challenging real-world scenario: a flexible target with a flat, water-exposed binding site. We identified a subset of 69 highly water-soluble MiniFrags that were screened against the antiapoptotic protein human Bfl-1.


Assuntos
Descoberta de Drogas , Bibliotecas de Moléculas Pequenas , Humanos , Peso Molecular , Bibliotecas de Moléculas Pequenas/química , Espectroscopia de Ressonância Magnética/métodos , Descoberta de Drogas/métodos , Cristalografia por Raios X , Água , Ligação Proteica , Ligantes
6.
J Mol Graph Model ; 129: 108734, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38442440

RESUMO

Application of Artificial intelligence (AI) in drug discovery has led to several success stories in recent times. While traditional methods mostly relied upon screening large chemical libraries for early-stage drug-design, de novo design can help identify novel target-specific molecules by sampling from a much larger chemical space. Although this has increased the possibility of finding diverse and novel molecules from previously unexplored chemical space, this has also posed a great challenge for medicinal chemists to synthesize at least some of the de novo designed novel molecules for experimental validation. To address this challenge, in this work, we propose a novel forward synthesis-based generative AI method, which is used to explore the synthesizable chemical space. The method uses a structure-based drug design framework, where the target protein structure and a target-specific seed fragment from co-crystal structures can be the initial inputs. A random fragment from a purchasable fragment library can also be the input if a target-specific fragment is unavailable. Then a template-based forward synthesis route prediction and molecule generation is performed in parallel using the Monte Carlo Tree Search (MCTS) method where, the subsequent fragments for molecule growth can again be obtained from a purchasable fragment library. The rewards for each iteration of MCTS are computed using a drug-target affinity (DTA) model based on the docking pose of the generated reaction intermediates at the binding site of the target protein of interest. With the help of the proposed method, it is now possible to overcome one of the major obstacles posed to the AI-based drug design approaches through the ability of the method to design novel target-specific synthesizable molecules.


Assuntos
Inteligência Artificial , Descoberta de Drogas , Descoberta de Drogas/métodos , Desenho de Fármacos , Proteínas/química , Bibliotecas de Moléculas Pequenas/química
7.
Int J Mol Sci ; 25(5)2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38473785

RESUMO

Deep learning is a machine learning technique to model high-level abstractions in data by utilizing a graph composed of multiple processing layers that experience various linear and non-linear transformations. This technique has been shown to perform well for applications in drug discovery, utilizing structural features of small molecules to predict activity. Here, we report a large-scale study to predict the activity of small molecules across the human kinome-a major family of drug targets, particularly in anti-cancer agents. While small-molecule kinase inhibitors exhibit impressive clinical efficacy in several different diseases, resistance often arises through adaptive kinome reprogramming or subpopulation diversity. Polypharmacology and combination therapies offer potential therapeutic strategies for patients with resistant diseases. Their development would benefit from a more comprehensive and dense knowledge of small-molecule inhibition across the human kinome. Leveraging over 650,000 bioactivity annotations for more than 300,000 small molecules, we evaluated multiple machine learning methods to predict the small-molecule inhibition of 342 kinases across the human kinome. Our results demonstrated that multi-task deep neural networks outperformed classical single-task methods, offering the potential for conducting large-scale virtual screening, predicting activity profiles, and bridging the gaps in the available data.


Assuntos
Aprendizado Profundo , Humanos , Fosfotransferases , Descoberta de Drogas/métodos , Polifarmacologia , Aprendizado de Máquina
8.
Commun Biol ; 7(1): 320, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38480979

RESUMO

Fragment screening is used to identify binding sites and leads in drug discovery, but it is often unclear which binding sites are functionally important. Here, data from 37 experiments, and 1309 protein structures binding to 1601 ligands were analysed. A method to group ligands by binding sites is introduced and sites clustered according to profiles of relative solvent accessibility. This identified 293 unique ligand binding sites, grouped into four clusters (C1-4). C1 includes larger, buried, conserved, and population missense-depleted sites, enriched in known functional sites. C4 comprises smaller, accessible, divergent, missense-enriched sites, depleted in functional sites. A site in C1 is 28 times more likely to be functional than one in C4. Seventeen sites, which to the best of our knowledge are novel, in 13 proteins are identified as likely to be functionally important with examples from human tenascin and 5-aminolevulinate synthase highlighted. A multi-layer perceptron, and K-nearest neighbours model are presented to predict cluster labels for ligand binding sites with an accuracy of 96% and 100%, respectively, so allowing functional classification of sites for proteins not in this set. Our findings will be of interest to those studying protein-ligand interactions and developing new drugs or function modulators.


Assuntos
Descoberta de Drogas , Proteínas , Humanos , Ligantes , Sítios de Ligação , Proteínas/metabolismo , Descoberta de Drogas/métodos
9.
J Comput Aided Mol Des ; 38(1): 13, 2024 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-38493240

RESUMO

The growing size of make-on-demand chemical libraries is posing new challenges to cheminformatics. These ultra-large chemical libraries became too large for exhaustive enumeration. Using a combinatorial approach instead, the resource requirement scales approximately with the number of synthons instead of the number of molecules. This gives access to billions or trillions of compounds as so-called chemical spaces with moderate hardware and in a reasonable time frame. While extremely performant ligand-based 2D methods exist in this context, 3D methods still largely rely on exhaustive enumeration and therefore fail to apply. Here, we present SpaceGrow: a novel shape-based 3D approach for ligand-based virtual screening of billions of compounds within hours on a single CPU. Compared to a conventional superposition tool, SpaceGrow shows comparable pose reproduction capacity based on RMSD and superior ranking performance while being orders of magnitude faster. Result assessment of two differently sized subsets of the eXplore space reveals a higher probability of finding superior results in larger spaces highlighting the potential of searching in ultra-large spaces. Furthermore, the application of SpaceGrow in a drug discovery workflow was investigated in four examples involving G protein-coupled receptors (GPCRs) with the aim to identify compounds with similar binding capabilities and molecular novelty.


Assuntos
Descoberta de Drogas , Bibliotecas de Moléculas Pequenas , Ligantes , Bibliotecas de Moléculas Pequenas/química , Descoberta de Drogas/métodos
10.
PLoS One ; 19(3): e0300906, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38512848

RESUMO

Given the prolonged timelines and high costs associated with traditional approaches, accelerating drug development is crucial. Computational methods, particularly drug-target interaction prediction, have emerged as efficient tools, yet the explainability of machine learning models remains a challenge. Our work aims to provide more interpretable interaction prediction models using similarity-based prediction in a latent space aligned to biological hierarchies. We investigated integrating drug and protein hierarchies into a joint-embedding drug-target latent space via embedding regularization by conducting a comparative analysis between models employing traditional flat Euclidean vector spaces and those utilizing hyperbolic embeddings. Besides, we provided a latent space analysis as an example to show how we can gain visual insights into the trained model with the help of dimensionality reduction. Our results demonstrate that hierarchy regularization improves interpretability without compromising predictive performance. Furthermore, integrating hyperbolic embeddings, coupled with regularization, enhances the quality of the embedded hierarchy trees. Our approach enables a more informed and insightful application of interaction prediction models in drug discovery by constructing an interpretable hyperbolic latent space, simultaneously incorporating drug and target hierarchies and pairing them with available interaction information. Moreover, compatible with pairwise methods, the approach allows for additional transparency through existing explainable AI solutions.


Assuntos
Descoberta de Drogas , Aprendizado de Máquina , Descoberta de Drogas/métodos , Desenvolvimento de Medicamentos , Proteínas
11.
J Chem Inf Model ; 64(6): 1882-1891, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38442000

RESUMO

Virtual screening of large compound libraries to identify potential hit candidates is one of the earliest steps in drug discovery. As the size of commercially available compound collections grows exponentially to the scale of billions, active learning and Bayesian optimization have recently been proven as effective methods of narrowing down the search space. An essential component of those methods is a surrogate machine learning model that predicts the desired properties of compounds. An accurate model can achieve high sample efficiency by finding hits with only a fraction of the entire library being virtually screened. In this study, we examined the performance of a pretrained transformer-based language model and graph neural network in a Bayesian optimization active learning framework. The best pretrained model identifies 58.97% of the top-50,000 compounds after screening only 0.6% of an ultralarge library containing 99.5 million compounds, improving 8% over the previous state-of-the-art baseline. Through extensive benchmarks, we show that the superior performance of pretrained models persists in both structure-based and ligand-based drug discovery. Pretrained models can serve as a boost to the accuracy and sample efficiency of active learning-based virtual screening.


Assuntos
Descoberta de Drogas , Bibliotecas de Moléculas Pequenas , Teorema de Bayes , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/química , Descoberta de Drogas/métodos , Redes Neurais de Computação , Aprendizado de Máquina
12.
J Chem Inf Model ; 64(6): 2084-2100, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38456842

RESUMO

The knowledge of ligand binding hot spots and of the important interactions within such hot spots is crucial for the design of lead compounds in the early stages of structure-based drug discovery. The computational solvent mapping server FTMap can reliably identify binding hot spots as consensus clusters, free energy minima that bind a variety of organic probe molecules. However, in its current implementation, FTMap provides limited information on regions within the hot spots that tend to interact with specific pharmacophoric features of potential ligands. E-FTMap is a new server that expands on the original FTMap protocol. E-FTMap uses 119 organic probes, rather than the 16 in the original FTMap, to exhaustively map binding sites, and identifies pharmacophore features as atomic consensus sites where similar chemical groups bind. We validate E-FTMap against a set of 109 experimentally derived structures of fragment-lead pairs, finding that highly ranked pharmacophore features overlap with the corresponding atoms in both fragments and lead compounds. Additionally, comparisons of mapping results to ensembles of bound ligands reveal that pharmacophores generated with E-FTMap tend to sample highly conserved protein-ligand interactions. E-FTMap is available as a web server at https://eftmap.bu.edu.


Assuntos
Descoberta de Drogas , Farmacóforo , Ligantes , Sítios de Ligação , Descoberta de Drogas/métodos , Ligação Proteica
13.
J Chem Inf Model ; 64(6): 1955-1965, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38446131

RESUMO

Active learning (AL) has become a powerful tool in computational drug discovery, enabling the identification of top binders from vast molecular libraries. To design a robust AL protocol, it is important to understand the influence of AL parameters, as well as the features of the data sets on the outcomes. We use four affinity data sets for different targets (TYK2, USP7, D2R, Mpro) to systematically evaluate the performance of machine learning models [Gaussian process (GP) model and Chemprop model], sample selection protocols, and the batch size based on metrics describing the overall predictive power of the model (R2, Spearman rank, root-mean-square error) as well as the accurate identification of top 2%/5% binders (Recall, F1 score). Both models have a comparable Recall of top binders on large data sets, but the GP model surpasses the Chemprop model when training data are sparse. A larger initial batch size, especially on diverse data sets, increased the Recall of both models as well as overall correlation metrics. However, for subsequent cycles, smaller batch sizes of 20 or 30 compounds proved to be desirable. Furthermore, adding artificial Gaussian noise to the data up to a certain threshold still allowed the model to identify clusters with top-scoring compounds. However, excessive noise (<1σ) did impact the model's predictive and exploitative capabilities.


Assuntos
Benchmarking , Aprendizado de Máquina , Ligantes , Descoberta de Drogas/métodos
14.
J Med Chem ; 67(6): 4322-4345, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38457829

RESUMO

Photochemistry has emerged as a transformative force in organic chemistry, significantly expanding the chemical space accessible for medicinal chemistry. Light-induced reactions enable the efficient synthesis of intricate organic structures and have found applications throughout the different stages of the drug discovery and development processes. Moreover, photochemical techniques provide innovative solutions in chemical biology, allowing precise spatiotemporal drug activation and targeted delivery. In this Perspective, we highlight the already numerous remarkable applications and the even more promising future of photochemistry in medicinal chemistry and chemical biology.


Assuntos
Química Farmacêutica , Descoberta de Drogas , Fotoquímica , Química Farmacêutica/métodos , Descoberta de Drogas/métodos , Biologia
15.
Math Biosci Eng ; 21(2): 2608-2625, 2024 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-38454698

RESUMO

In the drug discovery process, time and costs are the most typical problems resulting from the experimental screening of drug-target interactions (DTIs). To address these limitations, many computational methods have been developed to achieve more accurate predictions. However, identifying DTIs mostly rely on separate learning tasks with drug and target features that neglect interaction representation between drugs and target. In addition, the lack of these relationships may lead to a greatly impaired performance on the prediction of DTIs. Aiming at capturing comprehensive drug-target representations and simplifying the network structure, we propose an integrative approach with a convolution broad learning system for the DTI prediction (ConvBLS-DTI) to reduce the impact of the data sparsity and incompleteness. First, given the lack of known interactions for the drug and target, the weighted K-nearest known neighbors (WKNKN) method was used as a preprocessing strategy for unknown drug-target pairs. Second, a neighborhood regularized logistic matrix factorization (NRLMF) was applied to extract features of updated drug-target interaction information, which focused more on the known interaction pair parties. Then, a broad learning network incorporating a convolutional neural network was established to predict DTIs, which can make classification more effective using a different perspective. Finally, based on the four benchmark datasets in three scenarios, the ConvBLS-DTI's overall performance out-performed some mainstream methods. The test results demonstrate that our model achieves improved prediction effect on the area under the receiver operating characteristic curve and the precision-recall curve.


Assuntos
Descoberta de Drogas , Redes Neurais de Computação , Descoberta de Drogas/métodos , Curva ROC
16.
J Med Chem ; 67(7): 5683-5698, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38498697

RESUMO

Developing orally bioavailable drugs demands an understanding of absorption in early drug development. Traditional methods and physicochemical properties optimize absorption for rule of five (Ro5) compounds; beyond rule of five (bRo5) drugs necessitate advanced tools like the experimental measure of exposed polarity (EPSA) and the AbbVie multiparametric score (AB-MPS). Analyzing AB-MPS and EPSA against ∼1000 compounds with human absorption data and ∼10,000 AbbVie tool compounds (∼1000 proteolysis targeting chimeras or PROTACs, ∼7000 Ro5s, and ∼2000 bRo5s) revealed new patterns of physicochemical trends. We introduced a high-throughput "polarity reduction" descriptor: ETR, the EPSA-to-topological polar surface area (TPSA) ratio, highlights unique bRo5 and PROTAC subsets for specialized drug design strategies for effective absorption. Our methods and guidelines refine drug design by providing innovative in vitro approaches, enhancing physicochemical property optimization, and enabling accurate predictions of intestinal absorption in the complex bRo5 domain.


Assuntos
Descoberta de Drogas , Quimera de Direcionamento de Proteólise , Humanos , Descoberta de Drogas/métodos , Desenho de Fármacos , Absorção Intestinal , Proteólise
17.
Mol Pharm ; 21(4): 1563-1590, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38466810

RESUMO

Understanding protein sequence and structure is essential for understanding protein-protein interactions (PPIs), which are essential for many biological processes and diseases. Targeting protein binding hot spots, which regulate signaling and growth, with rational drug design is promising. Rational drug design uses structural data and computational tools to study protein binding sites and protein interfaces to design inhibitors that can change these interactions, thereby potentially leading to therapeutic approaches. Artificial intelligence (AI), such as machine learning (ML) and deep learning (DL), has advanced drug discovery and design by providing computational resources and methods. Quantum chemistry is essential for drug reactivity, toxicology, drug screening, and quantitative structure-activity relationship (QSAR) properties. This review discusses the methodologies and challenges of identifying and characterizing hot spots and binding sites. It also explores the strategies and applications of artificial-intelligence-based rational drug design technologies that target proteins and protein-protein interaction (PPI) binding hot spots. It provides valuable insights for drug design with therapeutic implications. We have also demonstrated the pathological conditions of heat shock protein 27 (HSP27) and matrix metallopoproteinases (MMP2 and MMP9) and designed inhibitors of these proteins using the drug discovery paradigm in a case study on the discovery of drug molecules for cancer treatment. Additionally, the implications of benzothiazole derivatives for anticancer drug design and discovery are deliberated.


Assuntos
Inteligência Artificial , Descoberta de Drogas , Descoberta de Drogas/métodos , Desenho de Fármacos , Aprendizado de Máquina , Relação Quantitativa Estrutura-Atividade
18.
J Chem Inf Model ; 64(4): 1158-1171, 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38316125

RESUMO

Over the last five years, virtual screening of ultralarge synthesis on-demand libraries has emerged as a powerful tool for hit identification in drug discovery programs. As these libraries have grown to tens of billions of molecules, we have reached a point where it is no longer cost-effective to screen every molecule virtually. To address these challenges, several groups have developed heuristic search methods to rapidly identify the best molecules on a virtual screen. This article describes the application of Thompson sampling (TS), an active learning approach that streamlines the virtual screening of large combinatorial libraries by performing a probabilistic search in the reagent space, thereby never requiring the full enumeration of the library. TS is a general technique that can be applied to various virtual screening modalities, including 2D and 3D similarity search, docking, and application of machine-learning models. In an illustrative example, we show that TS can identify more than half of the top 100 molecules from a docking-based virtual screen of 335 million molecules by evaluating 1% of the data set.


Assuntos
Bases de Dados de Compostos Químicos , Descoberta de Drogas , Descoberta de Drogas/métodos
19.
SLAS Discov ; 29(2): 100146, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38311110

RESUMO

Here we offer perspectives on phenotypic screening based on a wide-ranging discussion entitled "Phenotypic screening, target ID, and multi-omics: enabling more disease relevance in early discovery?" at the Screen Design and Assay Technology Special Interest Group Meeting at the 2023 SLAS Conference. During the session, the authors shared their own experience from within their respective organizations, followed by an open discussion with the audience. It was recognized that while substantial progress has been made towards translating disease-relevant phenotypic early discovery into clinical success, there remain significant operational and scientific challenges to implementing phenotypic screening efforts, and improving translation of screening hits comes with substantial resource demands and organizational commitment. This Perspective assesses progress, highlights pitfalls, and offers possible solutions to help unlock the therapeutic potential of phenotypic drug discovery. Areas explored comprise screening and hit validation strategy, choice of cellular model, moving beyond 2D cell culture into three dimensions, and leveraging high-dimensional data sets downstream of phenotypic screens.


Assuntos
Descoberta de Drogas , Opinião Pública , Descoberta de Drogas/métodos , Fenótipo
20.
Expert Opin Drug Discov ; 19(4): 415-431, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38321848

RESUMO

INTRODUCTION: Targeting RNAs with small molecules offers an alternative to the conventional protein-targeted drug discovery and can potentially address unmet and emerging medical needs. The recent rise of interest in the strategy has already resulted in large amounts of data on disease associated RNAs, as well as on small molecules that bind to such RNAs. Artificial intelligence (AI) approaches, including machine learning and deep learning, present an opportunity to speed up the discovery of RNA-targeted small molecules by improving decision-making efficiency and quality. AREAS COVERED: The topics described in this review include the recent applications of AI in the identification of RNA targets, RNA structure determination, screening of chemical compound libraries, and hit-to-lead optimization. The impact and limitations of the recent AI applications are discussed, along with an outlook on the possible applications of next-generation AI tools for the discovery of novel RNA-targeted small molecule drugs. EXPERT OPINION: Key areas for improvement include developing AI tools for understanding RNA dynamics and RNA - small molecule interactions. High-quality and comprehensive data still need to be generated especially on the biological activity of small molecules that target RNAs.


Assuntos
Inteligência Artificial , RNA , Humanos , Descoberta de Drogas/métodos , Aprendizado de Máquina , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...