Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38397627

RESUMEN

Most research on forest therapy has examined the therapeutic effects of forest activity development. There has been insufficient research identifying and evaluating the forest therapy environment. This study aimed to derive a representative forest therapy environment from each of the four evaluation sites, comprising national luxury forests; Scopus, PubMed, Medline, Web of Science, RISS, and DBpia were searched, and 13 studies evaluating forest therapy environments were analyzed and synthesized. After conducting a Conformity Evaluation, one layer of items, comprising anions with low conformity scores, was excluded, and six field measurements, phytoncide, oxygen, illuminance, UV-rays, sound, and anion, were added to increase objectivity. Finally, five forest therapy environment categories and 25 detailed items were derived. Analytic Hierarchy Process-based importance was evaluated to calculate the weight between the final evaluation items. According to the site evaluations, the categories of landscape, forest air, sunlight, sound, and anions appeared, in that order. This study is significant as it developed evaluation items and rating criteria for forest therapy environments, applied these in the field, and derived representative forest therapy environments for each location. This study developed indicators, provided basic data for establishing a therapy environment management plan, and there recommendations were made for an environment suitable for visitors and customizing forest welfare and therapy services.


Asunto(s)
Bosques , Oxígeno , Aniones
2.
J Am Med Inform Assoc ; 31(1): 35-44, 2023 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-37604111

RESUMEN

OBJECTIVE: Applications of machine learning in healthcare are of high interest and have the potential to improve patient care. Yet, the real-world accuracy of these models in clinical practice and on different patient subpopulations remains unclear. To address these important questions, we hosted a community challenge to evaluate methods that predict healthcare outcomes. We focused on the prediction of all-cause mortality as the community challenge question. MATERIALS AND METHODS: Using a Model-to-Data framework, 345 registered participants, coalescing into 25 independent teams, spread over 3 continents and 10 countries, generated 25 accurate models all trained on a dataset of over 1.1 million patients and evaluated on patients prospectively collected over a 1-year observation of a large health system. RESULTS: The top performing team achieved a final area under the receiver operator curve of 0.947 (95% CI, 0.942-0.951) and an area under the precision-recall curve of 0.487 (95% CI, 0.458-0.499) on a prospectively collected patient cohort. DISCUSSION: Post hoc analysis after the challenge revealed that models differ in accuracy on subpopulations, delineated by race or gender, even when they are trained on the same data. CONCLUSION: This is the largest community challenge focused on the evaluation of state-of-the-art machine learning methods in a healthcare system performed to date, revealing both opportunities and pitfalls of clinical AI.


Asunto(s)
Colaboración de las Masas , Medicina , Humanos , Inteligencia Artificial , Aprendizaje Automático , Algoritmos
3.
Database (Oxford) ; 20232023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-37551911

RESUMEN

Biomedical relation extraction (BioRE) is the task of automatically extracting and classifying relations between two biomedical entities in biomedical literature. Recent advances in BioRE research have largely been powered by supervised learning and large language models (LLMs). However, training of LLMs for BioRE with supervised learning requires human-annotated data, and the annotation process often accompanies challenging and expensive work. As a result, the quantity and coverage of annotated data are limiting factors for BioRE systems. In this paper, we present our system for the BioCreative VII challenge-DrugProt track, a BioRE system that leverages a language model structure and weak supervision. Our system is trained on weakly labelled data and then fine-tuned using human-labelled data. To create the weakly labelled dataset, we combined two approaches. First, we trained a model on the original dataset to predict labels on external literature, which will become a model-labelled dataset. Then, we refined the model-labelled dataset using an external knowledge base. Based on our experiment, our approach using refined weak supervision showed significant performance gain over the model trained using standard human-labelled datasets. Our final model showed outstanding performance at the BioCreative VII challenge, achieving 3rd place (this paper focuses on our participating system in the BioCreative VII challenge). Database URL: http://wonjin.info/biore-yoon-et-al-2022.


Asunto(s)
Investigación Biomédica , Lenguaje , Humanos , Bases de Datos Factuales
4.
Bioinformatics ; 39(39 Suppl 1): i448-i457, 2023 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-37387164

RESUMEN

MOTIVATION: Protein-ligand binding affinity prediction is a central task in drug design and development. Cross-modal attention mechanism has recently become a core component of many deep learning models due to its potential to improve model explainability. Non-covalent interactions (NCIs), one of the most critical domain knowledge in binding affinity prediction task, should be incorporated into protein-ligand attention mechanism for more explainable deep drug-target interaction models. We propose ArkDTA, a novel deep neural architecture for explainable binding affinity prediction guided by NCIs. RESULTS: Experimental results show that ArkDTA achieves predictive performance comparable to current state-of-the-art models while significantly improving model explainability. Qualitative investigation into our novel attention mechanism reveals that ArkDTA can identify potential regions for NCIs between candidate drug compounds and target proteins, as well as guiding internal operations of the model in a more interpretable and domain-aware manner. AVAILABILITY: ArkDTA is available at https://github.com/dmis-lab/ArkDTA. CONTACT: kangj@korea.ac.kr.


Asunto(s)
Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Ligandos
5.
Bioinformatics ; 39(6)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37261870

RESUMEN

SUMMARY: Biomedical named entity recognition (NER) plays a crucial role in extracting information from documents in biomedical applications. However, many of these applications require NER models to operate at a document level, rather than just a sentence level. This presents a challenge, as the extension from a sentence model to a document model is not always straightforward. Despite the existence of document NER models that are able to make consistent predictions, they still fall short of meeting the expectations of researchers and practitioners in the field. To address this issue, we have undertaken an investigation into the underlying causes of inconsistent predictions. Our research has led us to believe that the use of adjectives and prepositions within entities may be contributing to low label consistency. In this article, we present our method, ConNER, to enhance a label consistency of modifiers such as adjectives and prepositions. By refining the labels of these modifiers, ConNER is able to improve representations of biomedical entities. The effectiveness of our method is demonstrated on four popular biomedical NER datasets. On three datasets, we achieve a higher F1 score than the previous state-of-the-art model. Our method shows its efficacy on two datasets, resulting in 7.5%-8.6% absolute improvements in the F1 score. Our findings suggest that our ConNER method is effective on datasets with intrinsically low label consistency. Through qualitative analysis, we demonstrate how our approach helps the NER model generate more consistent predictions. AVAILABILITY AND IMPLEMENTATION: Our code and resources are available at https://github.com/dmis-lab/ConNER/.


Asunto(s)
Minería de Datos , Lenguaje , Humanos , Minería de Datos/métodos , Investigadores
6.
Bioinformatics ; 39(1)2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36416124

RESUMEN

MOTIVATION: Compound-protein interaction (CPI) plays an essential role in drug discovery and is performed via expensive molecular docking simulations. Many artificial intelligence-based approaches have been proposed in this regard. Recently, two types of models have accomplished promising results in exploiting molecular information: graph convolutional neural networks that construct a learned molecular representation from a graph structure (atoms and bonds), and neural networks that can be applied to compute on descriptors or fingerprints of molecules. However, the superiority of one method over the other is yet to be determined. Modern studies have endeavored to aggregate information that is extracted from compounds and proteins to form the CPI task. Nonetheless, these approaches have used a simple concatenation to combine them, which cannot fully capture the interaction between such information. RESULTS: We propose the Perceiver CPI network, which adopts a cross-attention mechanism to improve the learning ability of the representation of drug and target interactions and exploits the rich information obtained from extended-connectivity fingerprints to improve the performance. We evaluated Perceiver CPI on three main datasets, Davis, KIBA and Metz, to compare the performance of our proposed model with that of state-of-the-art methods. The proposed method achieved satisfactory performance and exhibited significant improvements over previous approaches in all experiments. AVAILABILITY AND IMPLEMENTATION: Perceiver CPI is available at https://github.com/dmis-lab/PerceiverCPI. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Inteligencia Artificial , Redes Neurales de la Computación , Simulación del Acoplamiento Molecular , Proteínas/química , Mapas de Interacción de Proteínas
7.
Brief Bioinform ; 25(1)2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-38180829

RESUMEN

Forecasting the interaction between compounds and proteins is crucial for discovering new drugs. However, previous sequence-based studies have not utilized three-dimensional (3D) information on compounds and proteins, such as atom coordinates and distance matrices, to predict binding affinity. Furthermore, numerous widely adopted computational techniques have relied on sequences of amino acid characters for protein representations. This approach may constrain the model's ability to capture meaningful biochemical features, impeding a more comprehensive understanding of the underlying proteins. Here, we propose a two-step deep learning strategy named MulinforCPI that incorporates transfer learning techniques with multi-level resolution features to overcome these limitations. Our approach leverages 3D information from both proteins and compounds and acquires a profound understanding of the atomic-level features of proteins. Besides, our research highlights the divide between first-principle and data-driven methods, offering new research prospects for compound-protein interaction tasks. We applied the proposed method to six datasets: Davis, Metz, KIBA, CASF-2016, DUD-E and BindingDB, to evaluate the effectiveness of our approach.


Asunto(s)
Aminoácidos , Mapeo de Interacción de Proteínas , Conformación Proteica , Unión Proteica
8.
Sci Rep ; 12(1): 19636, 2022 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-36385263

RESUMEN

Association between exposure to periodontal bacteria and development of autoantibodies related to rheumatoid arthritis (RA) has been widely accepted; however, direct causal relationship between periodontal bacteria and rheumatoid factor (RF) is currently not fully understood. We investigated whether periodontal bacteria could affect RF status. Patients with preclinical, new-onset, or chronic RA underwent periodontal examination, and investigation of subgingival microbiome via 16S rRNA sequencing. Degree of arthritis and RF induction was examined in collagen-induced arthritis (CIA) mice that were orally inoculated with different periodontal bacteria species. Subsequently, single-cell RNA sequencing analysis of the mouse spleen cells was performed. Patients with preclinical RA showed an increased abundance of the Porphyromonadacae family in the subgingival microbiome compared to those with new-onset or chronic RA, despite comparable periodontitis severity among them. Notably, a distinct subgingival microbial community was found between patients with high-positive RF and those with negative or low-positive RF (p=0.022). Oral infections with the periodontal pathogens P. gingivalis and Treponema denticola in CIA mice similarly enhanced arthritis score, but resulted in different levels of RF induction. Genes related to B cell receptor signaling, B cell proliferation, activation, and differentiation, and CD4+ T cell costimulation and cytokine production were involved in the differential induction of RF in mice exposed to different bacteria. In summary, periodontal microbiome might shape RF status by affecting the humoral immune response during RA pathogenesis.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Microbiota , Ratones , Animales , Factor Reumatoide , ARN Ribosómico 16S/genética , Microbiota/genética , Treponema denticola
9.
Bioinformatics ; 38(20): 4837-4839, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36053172

RESUMEN

In biomedical natural language processing, named entity recognition (NER) and named entity normalization (NEN) are key tasks that enable the automatic extraction of biomedical entities (e.g. diseases and drugs) from the ever-growing biomedical literature. In this article, we present BERN2 (Advanced Biomedical Entity Recognition and Normalization), a tool that improves the previous neural network-based NER tool by employing a multi-task NER model and neural network-based NEN models to achieve much faster and more accurate inference. We hope that our tool can help annotate large-scale biomedical texts for various tasks such as biomedical knowledge graph construction. AVAILABILITY AND IMPLEMENTATION: Web service of BERN2 is publicly available at http://bern2.korea.ac.kr. We also provide local installation of BERN2 at https://github.com/dmis-lab/BERN2. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Redes Neurales de la Computación , Programas Informáticos , Procesamiento de Lenguaje Natural
10.
Database (Oxford) ; 20222022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36170114

RESUMEN

Chemical identification involves finding chemical entities in text (i.e. named entity recognition) and assigning unique identifiers to the entities (i.e. named entity normalization). While current models are developed and evaluated based on article titles and abstracts, their effectiveness has not been thoroughly verified in full text. In this paper, we identify two limitations of models in tagging full-text articles: (1) low generalizability to unseen mentions and (2) tagging inconsistency. We use simple training and post-processing methods to address the limitations such as transfer learning and mention-wise majority voting. We also present a hybrid model for the normalization task that utilizes the high recall of a neural model while maintaining the high precision of a dictionary model. In the BioCreative VII NLM-Chem track challenge, our best model achieves 86.72 and 78.31 F1 scores in named entity recognition and normalization, significantly outperforming the median (83.73 and 77.49 F1 scores) and taking first place in named entity recognition. In a post-challenge evaluation, we re-implement our model and obtain 84.70 F1 score in the normalization task, outperforming the best score in the challenge by 3.34 F1 score. Database URL: https://github.com/dmis-lab/bc7-chem-id.


Asunto(s)
Minería de Datos , Minería de Datos/métodos , Bases de Datos Factuales
11.
Neural Netw ; 153: 104-119, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35716619

RESUMEN

Graph Neural Networks (GNNs) have been widely applied to various fields due to their powerful representations of graph-structured data. Despite the success of GNNs, most existing GNNs are designed to learn node representations on the fixed and homogeneous graphs. The limitations especially become problematic when learning representations on a misspecified graph or a heterogeneous graph that consists of various types of nodes and edges. To address these limitations, we propose Graph Transformer Networks (GTNs) that are capable of generating new graph structures, which preclude noisy connections and include useful connections (e.g., meta-paths) for tasks, while learning effective node representations on the new graphs in an end-to-end fashion. We further propose enhanced version of GTNs, Fast Graph Transformer Networks (FastGTNs), that improve scalability of graph transformations. Compared to GTNs, FastGTNs are up to 230× and 150× faster in inference and training, and use up to 100× and 148× less memory while allowing the identical graph transformations as GTNs. In addition, we extend graph transformations to the semantic proximity of nodes allowing non-local operations beyond meta-paths. Extensive experiments on both homogeneous graphs and heterogeneous graphs show that GTNs and FastGTNs with non-local operations achieve the state-of-the-art performance for node classification tasks. The code is available: https://github.com/seongjunyun/Graph_Transformer_Networks.


Asunto(s)
Aprendizaje , Redes Neurales de la Computación , Semántica
12.
Bioinformatics ; 38(15): 3794-3801, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35713500

RESUMEN

MOTIVATION: Current studies in extractive question answering (EQA) have modeled the single-span extraction setting, where a single answer span is a label to predict for a given question-passage pair. This setting is natural for general domain EQA as the majority of the questions in the general domain can be answered with a single span. Following general domain EQA models, current biomedical EQA (BioEQA) models utilize the single-span extraction setting with post-processing steps. RESULTS: In this article, we investigate the question distribution across the general and biomedical domains and discover biomedical questions are more likely to require list-type answers (multiple answers) than factoid-type answers (single answer). This necessitates the models capable of producing multiple answers for a question. Based on this preliminary study, we propose a sequence tagging approach for BioEQA, which is a multi-span extraction setting. Our approach directly tackles questions with a variable number of phrases as their answer and can learn to decide the number of answers for a question from training data. Our experimental results on the BioASQ 7b and 8b list-type questions outperformed the best-performing existing models without requiring post-processing steps. AVAILABILITY AND IMPLEMENTATION: Source codes and resources are freely available for download at https://github.com/dmis-lab/SeqTagQA. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Biología Computacional , Programas Informáticos
13.
IEEE Access ; 10: 31513-31523, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35582496

RESUMEN

The number of biomedical literature on new biomedical concepts is rapidly increasing, which necessitates a reliable biomedical named entity recognition (BioNER) model for identifying new and unseen entity mentions. However, it is questionable whether existing models can effectively handle them. In this work, we systematically analyze the three types of recognition abilities of BioNER models: memorization, synonym generalization, and concept generalization. We find that although current best models achieve state-of-the-art performance on benchmarks based on overall performance, they have limitations in identifying synonyms and new biomedical concepts, indicating they are overestimated in terms of their generalization abilities. We also investigate failure cases of models and identify several difficulties in recognizing unseen mentions in biomedical literature as follows: (1) models tend to exploit dataset biases, which hinders the models' abilities to generalize, and (2) several biomedical names have novel morphological patterns with weak name regularity, and models fail to recognize them. We apply a statistics-based debiasing method to our problem as a simple remedy and show the improvement in generalization to unseen mentions. We hope that our analyses and findings would be able to facilitate further research into the generalization capabilities of NER models in a domain where their reliability is of utmost importance.

14.
J Assoc Inf Sci Technol ; 73(8): 1065-1078, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35441082

RESUMEN

Scientific novelty drives the efforts to invent new vaccines and solutions during the pandemic. First-time collaboration and international collaboration are two pivotal channels to expand teams' search activities for a broader scope of resources required to address the global challenge, which might facilitate the generation of novel ideas. Our analysis of 98,981 coronavirus papers suggests that scientific novelty measured by the BioBERT model that is pretrained on 29 million PubMed articles, and first-time collaboration increased after the outbreak of COVID-19, and international collaboration witnessed a sudden decrease. During COVID-19, papers with more first-time collaboration were found to be more novel and international collaboration did not hamper novelty as it had done in the normal periods. The findings suggest the necessity of reaching out for distant resources and the importance of maintaining a collaborative scientific community beyond nationalism during a pandemic.

15.
Front Oncol ; 11: 747250, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868947

RESUMEN

Most electronic medical records, such as free-text radiological reports, are unstructured; however, the methodological approaches to analyzing these accumulating unstructured records are limited. This article proposes a deep-transfer-learning-based natural language processing model that analyzes serial magnetic resonance imaging reports of rectal cancer patients and predicts their overall survival. To evaluate the model, a retrospective cohort study of 4,338 rectal cancer patients was conducted. The experimental results revealed that the proposed model utilizing pre-trained clinical linguistic knowledge could predict the overall survival of patients without any structured information and was superior to the carcinoembryonic antigen in predicting survival. The deep-transfer-learning model using free-text radiological reports can predict the survival of patients with rectal cancer, thereby increasing the utility of unstructured medical big data.

16.
PLoS Comput Biol ; 17(9): e1009302, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34520464

RESUMEN

A continuing challenge in modern medicine is the identification of safer and more efficacious drugs. Precision therapeutics, which have one molecular target, have been long promised to be safer and more effective than traditional therapies. This approach has proven to be challenging for multiple reasons including lack of efficacy, rapidly acquired drug resistance, and narrow patient eligibility criteria. An alternative approach is the development of drugs that address the overall disease network by targeting multiple biological targets ('polypharmacology'). Rational development of these molecules will require improved methods for predicting single chemical structures that target multiple drug targets. To address this need, we developed the Multi-Targeting Drug DREAM Challenge, in which we challenged participants to predict single chemical entities that target pro-targets but avoid anti-targets for two unrelated diseases: RET-based tumors and a common form of inherited Tauopathy. Here, we report the results of this DREAM Challenge and the development of two neural network-based machine learning approaches that were applied to the challenge of rational polypharmacology. Together, these platforms provide a potentially useful first step towards developing lead therapeutic compounds that address disease complexity through rational polypharmacology.


Asunto(s)
Desarrollo de Medicamentos , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-ret/antagonistas & inhibidores , Tauopatías/tratamiento farmacológico , Humanos , Neoplasias/metabolismo , Redes Neurales de la Computación , Polifarmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
17.
Int J Mol Sci ; 22(16)2021 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-34445802

RESUMEN

Osteoporosis is commonly treated via the long-term usage of anti-osteoporotic agents; however, poor drug compliance and undesirable side effects limit their treatment efficacy. The parathyroid hormone-related protein (PTHrP) is essential for normal bone formation and remodeling; thus, may be used as an anti-osteoporotic agent. Here, we developed a platform for the delivery of a single peptide composed of two regions of the PTHrP protein (1-34 and 107-139); mcPTHrP 1-34+107-139 using a minicircle vector. We also transfected mcPTHrP 1-34+107-139 into human mesenchymal stem cells (MSCs) and generated Thru 1-34+107-139-producing engineered MSCs (eMSCs) as an alternative delivery system. Osteoporosis was induced in 12-week-old C57BL/6 female mice via ovariectomy. The ovariectomized (OVX) mice were then treated with the two systems; (1) mcPTHrP 1-34+107-139 was intravenously administered three times (once per week); (2) eMSCs were intraperitoneally administered twice (on weeks four and six). Compared with the control OVX mice, the mcPTHrP 1-34+107-139-treated group showed better trabecular bone structure quality, increased bone formation, and decreased bone resorption. Similar results were observed in the eMSCs-treated OVX mice. Altogether, these results provide experimental evidence to support the potential of delivering PTHrP 1-34+107-139 using the minicircle technology for the treatment of osteoporosis.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , ADN/administración & dosificación , Osteogénesis/efectos de los fármacos , Proteína Relacionada con la Hormona Paratiroidea/administración & dosificación , Animales , Densidad Ósea/efectos de los fármacos , Línea Celular , Femenino , Células HEK293 , Humanos , Inyecciones Intravenosas/métodos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Osteoporosis/tratamiento farmacológico , Ovariectomía/métodos
18.
Bioinformatics ; 37(Suppl_1): i376-i382, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34252937

RESUMEN

MOTIVATION: Identifying mechanism of actions (MoA) of novel compounds is crucial in drug discovery. Careful understanding of MoA can avoid potential side effects of drug candidates. Efforts have been made to identify MoA using the transcriptomic signatures induced by compounds. However, these approaches fail to reveal MoAs in the absence of actual compound signatures. RESULTS: We present MoAble, which predicts MoAs without requiring compound signatures. We train a deep learning-based coembedding model to map compound signatures and compound structure into the same embedding space. The model generates low-dimensional compound signature representation from the compound structures. To predict MoAs, pathway enrichment analysis is performed based on the connectivity between embedding vectors of compounds and those of genetic perturbation. Results show that MoAble is comparable to the methods that use actual compound signatures. We demonstrate that MoAble can be used to reveal MoAs of novel compounds without measuring compound signatures with the same prediction accuracy as that with measuring them. AVAILABILITY AND IMPLEMENTATION: MoAble is available at https://github.com/dmis-lab/moable. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Programas Informáticos , Transcriptoma , Descubrimiento de Drogas
19.
Nat Commun ; 12(1): 3307, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34083538

RESUMEN

Despite decades of intensive search for compounds that modulate the activity of particular protein targets, a large proportion of the human kinome remains as yet undrugged. Effective approaches are therefore required to map the massive space of unexplored compound-kinase interactions for novel and potent activities. Here, we carry out a crowdsourced benchmarking of predictive algorithms for kinase inhibitor potencies across multiple kinase families tested on unpublished bioactivity data. We find the top-performing predictions are based on various models, including kernel learning, gradient boosting and deep learning, and their ensemble leads to a predictive accuracy exceeding that of single-dose kinase activity assays. We design experiments based on the model predictions and identify unexpected activities even for under-studied kinases, thereby accelerating experimental mapping efforts. The open-source prediction algorithms together with the bioactivities between 95 compounds and 295 kinases provide a resource for benchmarking prediction algorithms and for extending the druggable kinome.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Algoritmos , Benchmarking , Colaboración de las Masas , Bases de Datos Farmacéuticas , Aprendizaje Profundo , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Humanos , Cinética , Aprendizaje Automático , Modelos Biológicos , Modelos Químicos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Quinasas/química , Proteómica , Análisis de Regresión
20.
Neural Netw ; 140: 27-38, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33744711

RESUMEN

Although neural models have performed impressively well on various tasks such as image recognition and question answering, their reasoning ability has been measured in only few studies. In this work, we focus on spatial reasoning and explore the spatial understanding of neural models. First, we describe the following two spatial reasoning IQ tests: rotation and shape composition. Using well-defined rules, we constructed datasets that consist of various complexity levels. We designed a variety of experiments in terms of generalization, and evaluated six different baseline models on the newly generated datasets. We provide an analysis of the results and factors that affect the generalization abilities of models. Also, we analyze how neural models solve spatial reasoning tests with visual aids. We hope that our work can encourage further research into human-level spatial reasoning and provide a new direction for future work.


Asunto(s)
Aprendizaje Automático , Modelos Neurológicos , Navegación Espacial , Humanos , Solución de Problemas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA