We introduce a convolutional neural network that operates directly on graphs. These networks allow end-to-end learning of prediction pipelines whose inputs are graphs of arbitrary size and shape. The architecture we present generalizes standard molecular feature extraction methods based on circular fingerprints. We show that these data-driven features are more interpretable, and have better predictive performance on a variety of tasks.
translated by 谷歌翻译
Molecular "fingerprints" encoding structural information are the workhorse of cheminformatics and machine learning in drug discovery applications. However, fingerprint representations necessarily emphasize particular aspects of the molecular structure while ignoring others, rather than allowing the model to make datadriven decisions. We describe molecular graph convolutions, a machine learning architecture for learning from undirected graphs, specifically small molecules. Graph convolutions use a simple encoding of the molecular graph-atoms, bonds, distances, etc.-which allows the model to take greater advantage of information in the graph structure. Although graph convolutions do not outperform all fingerprint-based methods, they (along with other graph-based methods) represent a new paradigm in ligand-based virtual screening with exciting opportunities for future improvement.
translated by 谷歌翻译
图形神经网络(GNN)正在化学工程中出现,以基于分子图的物理化学特性端到端学习。 GNNS的一个关键要素是合并函数,将原子矢量结合到分子指纹中。大多数以前的作品都使用标准池功能来预测各种属性。但是,不合适的合并功能会导致概括不佳的非物理GNN。我们根据有关学习特性的物理知识比较并选择有意义的GNN合并方法。通过量子机械计算计算出的分子特性证明了物理池函数的影响。我们还将结果与最近的SET2Set合并方法进行了比较。我们建议使用总和池来预测取决于分子大小的性能并比较分子大小无关的属性的池函数。总体而言,我们表明物理池功能的使用显着增强了概括。
translated by 谷歌翻译
Graph classification is an important area in both modern research and industry. Multiple applications, especially in chemistry and novel drug discovery, encourage rapid development of machine learning models in this area. To keep up with the pace of new research, proper experimental design, fair evaluation, and independent benchmarks are essential. Design of strong baselines is an indispensable element of such works. In this thesis, we explore multiple approaches to graph classification. We focus on Graph Neural Networks (GNNs), which emerged as a de facto standard deep learning technique for graph representation learning. Classical approaches, such as graph descriptors and molecular fingerprints, are also addressed. We design fair evaluation experimental protocol and choose proper datasets collection. This allows us to perform numerous experiments and rigorously analyze modern approaches. We arrive to many conclusions, which shed new light on performance and quality of novel algorithms. We investigate application of Jumping Knowledge GNN architecture to graph classification, which proves to be an efficient tool for improving base graph neural network architectures. Multiple improvements to baseline models are also proposed and experimentally verified, which constitutes an important contribution to the field of fair model comparison.
translated by 谷歌翻译
Extended-connectivity fingerprints (ECFPs) are a novel class of topological fingerprints for molecular characterization. Historically, topological fingerprints were developed for substructure and similarity searching. ECFPs were developed specifically for structure-activity modeling. ECFPs are circular fingerprints with a number of useful qualities: they can be very rapidly calculated; they are not predefined and can represent an essentially infinite number of different molecular features (including stereochemical information); their features represent the presence of particular substructures, allowing easier interpretation of analysis results; and the ECFP algorithm can be tailored to generate different types of circular fingerprints, optimized for different uses. While the use of ECFPs has been widely adopted and validated, a description of their implementation has not previously been presented in the literature.
translated by 谷歌翻译
We seek to automate the design of molecules based on specific chemical properties. In computational terms, this task involves continuous embedding and generation of molecular graphs. Our primary contribution is the direct realization of molecular graphs, a task previously approached by generating linear SMILES strings instead of graphs. Our junction tree variational autoencoder generates molecular graphs in two phases, by first generating a tree-structured scaffold over chemical substructures, and then combining them into a molecule with a graph message passing network. This approach allows us to incrementally expand molecules while maintaining chemical validity at every step. We evaluate our model on multiple tasks ranging from molecular generation to optimization. Across these tasks, our model outperforms previous state-of-the-art baselines by a significant margin.
translated by 谷歌翻译
近年来,基于Weisfeiler-Leman算法的算法和神经架构,是一个众所周知的Graph同构问题的启发式问题,它成为具有图形和关系数据的机器学习的强大工具。在这里,我们全面概述了机器学习设置中的算法的使用,专注于监督的制度。我们讨论了理论背景,展示了如何将其用于监督的图形和节点表示学习,讨论最近的扩展,并概述算法的连接(置换 - )方面的神经结构。此外,我们概述了当前的应用和未来方向,以刺激进一步的研究。
translated by 谷歌翻译
Models that accurately predict properties based on chemical structure are valuable tools in drug discovery. However, for many properties, public and private training sets are typically small, and it is difficult for the models to generalize well outside of the training data. Recently, large language models have addressed this problem by using self-supervised pretraining on large unlabeled datasets, followed by fine-tuning on smaller, labeled datasets. In this paper, we report MolE, a molecular foundation model that adapts the DeBERTa architecture to be used on molecular graphs together with a two-step pretraining strategy. The first step of pretraining is a self-supervised approach focused on learning chemical structures, and the second step is a massive multi-task approach to learn biological information. We show that fine-tuning pretrained MolE achieves state-of-the-art results on 9 of the 22 ADMET tasks included in the Therapeutic Data Commons.
translated by 谷歌翻译
基于深度学习的分子建模的最新进步令人兴奋地加速硅药发现。可获得血清的生成模型,构建原子原子和键合或逐片键的分子。然而,许多药物发现项目需要固定的支架以存在于所生成的分子中,并纳入该约束仅探讨了该约束。在这里,我们提出了一种基于图形的模型,其自然地支持支架作为生成过程的初始种子,这是可能的,因为它不调节在发电历史上。我们的实验表明,Moler与最先进的方法进行了相当的方法,在无约会的分子优化任务上,并且在基于脚手架的任务上优于它们,而不是比现有方法从培训和样本更快的数量级。此外,我们展示了许多看似小设计选择对整体性能的影响。
translated by 谷歌翻译
Supervised learning on molecules has incredible potential to be useful in chemistry, drug discovery, and materials science. Luckily, several promising and closely related neural network models invariant to molecular symmetries have already been described in the literature. These models learn a message passing algorithm and aggregation procedure to compute a function of their entire input graph. At this point, the next step is to find a particularly effective variant of this general approach and apply it to chemical prediction benchmarks until we either solve them or reach the limits of the approach. In this paper, we reformulate existing models into a single common framework we call Message Passing Neural Networks (MPNNs) and explore additional novel variations within this framework. Using MPNNs we demonstrate state of the art results on an important molecular property prediction benchmark; these results are strong enough that we believe future work should focus on datasets with larger molecules or more accurate ground truth labels.Recently, large scale quantum chemistry calculation and molecular dynamics simulations coupled with advances in high throughput experiments have begun to generate data at an unprecedented rate. Most classical techniques do not make effective use of the larger amounts of data that are now available. The time is ripe to apply more powerful and flexible machine learning methods to these problems, assuming we can find models with suitable inductive biases. The symmetries of atomic systems suggest neural networks that operate on graph structured data and are invariant to graph isomorphism might also be appropriate for molecules. Sufficiently successful models could someday help automate challenging chemical search problems in drug discovery or materials science.In this paper, our goal is to demonstrate effective machine learning models for chemical prediction problems
translated by 谷歌翻译
人工智能(AI)在过去十年中一直在改变药物发现的实践。各种AI技术已在广泛的应用中使用,例如虚拟筛选和药物设计。在本调查中,我们首先概述了药物发现,并讨论了相关的应用,可以减少到两个主要任务,即分子性质预测和分子产生。然后,我们讨论常见的数据资源,分子表示和基准平台。此外,为了总结AI在药物发现中的进展情况,我们介绍了在调查的论文中包括模型架构和学习范式的相关AI技术。我们预计本调查将作为有兴趣在人工智能和药物发现界面工作的研究人员的指南。我们还提供了GitHub存储库(HTTPS:///github.com/dengjianyuan/survey_survey_au_drug_discovery),其中包含文件和代码,如适用,作为定期更新的学习资源。
translated by 谷歌翻译
电子密度$ \ rho(\ vec {r})$是用密度泛函理论(dft)计算地面能量的基本变量。除了总能量之外,$ \ rho(\ vec {r})$分布和$ \ rho(\ vec {r})$的功能通常用于捕获电子规模以功能材料和分子中的关键物理化学现象。方法提供对$ \ rho(\ vec {r})的可紊乱系统,其具有少量计算成本的复杂无序系统可以是对材料相位空间的加快探索朝向具有更好功能的新材料的逆设计的游戏更换者。我们为预测$ \ rho(\ vec {r})$。该模型基于成本图形神经网络,并且在作为消息传递图的一部分的特殊查询点顶点上预测了电子密度,但仅接收消息。该模型在多个数据组中进行测试,分子(QM9),液体乙烯碳酸酯电解质(EC)和Lixniymnzco(1-Y-Z)O 2锂离子电池阴极(NMC)。对于QM9分子,所提出的模型的准确性超过了从DFT获得的$ \ Rho(\ vec {r})$中的典型变异性,以不同的交换相关功能,并显示超出最先进的准确性。混合氧化物(NMC)和电解质(EC)数据集更好的精度甚至更好。线性缩放模型同时探测成千上万点的能力允许计算$ \ Rho(\ vec {r})$的大型复杂系统,比DFT快于允许筛选无序的功能材料。
translated by 谷歌翻译
Advancements in neural machinery have led to a wide range of algorithmic solutions for molecular property prediction. Two classes of models in particular have yielded promising results: neural networks applied to computed molecular fingerprints or expert-crafted descriptors, and graph convolutional neural networks that construct a learned molecular representation by operating on the graph structure of the molecule.However, recent literature has yet to clearly determine which of these two methods is superior when generalizing to new chemical space. Furthermore, prior research has
translated by 谷歌翻译
The accurate prediction of physicochemical properties of chemical compounds in mixtures (such as the activity coefficient at infinite dilution $\gamma_{ij}^\infty$) is essential for developing novel and more sustainable chemical processes. In this work, we analyze the performance of previously-proposed GNN-based models for the prediction of $\gamma_{ij}^\infty$, and compare them with several mechanistic models in a series of 9 isothermal studies. Moreover, we develop the Gibbs-Helmholtz Graph Neural Network (GH-GNN) model for predicting $\ln \gamma_{ij}^\infty$ of molecular systems at different temperatures. Our method combines the simplicity of a Gibbs-Helmholtz-derived expression with a series of graph neural networks that incorporate explicit molecular and intermolecular descriptors for capturing dispersion and hydrogen bonding effects. We have trained this model using experimentally determined $\ln \gamma_{ij}^\infty$ data of 40,219 binary-systems involving 1032 solutes and 866 solvents, overall showing superior performance compared to the popular UNIFAC-Dortmund model. We analyze the performance of GH-GNN for continuous and discrete inter/extrapolation and give indications for the model's applicability domain and expected accuracy. In general, GH-GNN is able to produce accurate predictions for extrapolated binary-systems if at least 25 systems with the same combination of solute-solvent chemical classes are contained in the training set and a similarity indicator above 0.35 is also present. This model and its applicability domain recommendations have been made open-source at https://github.com/edgarsmdn/GH-GNN.
translated by 谷歌翻译
In this work, we propose MEDICO, a Multi-viEw Deep generative model for molecule generation, structural optimization, and the SARS-CoV-2 Inhibitor disCOvery. To the best of our knowledge, MEDICO is the first-of-this-kind graph generative model that can generate molecular graphs similar to the structure of targeted molecules, with a multi-view representation learning framework to sufficiently and adaptively learn comprehensive structural semantics from targeted molecular topology and geometry. We show that our MEDICO significantly outperforms the state-of-the-art methods in generating valid, unique, and novel molecules under benchmarking comparisons. In particular, we showcase the multi-view deep learning model enables us to generate not only the molecules structurally similar to the targeted molecules but also the molecules with desired chemical properties, demonstrating the strong capability of our model in exploring the chemical space deeply. Moreover, case study results on targeted molecule generation for the SARS-CoV-2 main protease (Mpro) show that by integrating molecule docking into our model as chemical priori, we successfully generate new small molecules with desired drug-like properties for the Mpro, potentially accelerating the de novo design of Covid-19 drugs. Further, we apply MEDICO to the structural optimization of three well-known Mpro inhibitors (N3, 11a, and GC376) and achieve ~88% improvement in their binding affinity to Mpro, demonstrating the application value of our model for the development of therapeutics for SARS-CoV-2 infection.
translated by 谷歌翻译
Molecular machine learning has been maturing rapidly over the last few years.Improved methods and the presence of larger datasets have enabled machine learning algorithms to make increasingly accurate predictions about molecular properties. However, algorithmic progress has been limited due to the lack of a standard benchmark to compare the efficacy of proposed methods; most new algorithms are benchmarked on different datasets making it challenging to gauge the quality of proposed methods. This work introduces MoleculeNet, a large scale benchmark for molecular machine learning. MoleculeNet curates multiple public datasets, establishes metrics for evaluation, and offers high quality open-source implementations of multiple previously proposed molecular featurization and learning algorithms (released as part of the DeepChem
translated by 谷歌翻译
在药物发现中,分子优化是在所需药物性质方面将药物候选改变为更好的阶梯。随着近期人工智能的进展,传统上的体外过程越来越促进了Silico方法。我们以硅方法提出了一种创新的,以通过深生成模型制定分子并制定问题,以便产生优化的分子图。我们的生成模型遵循基于片段的药物设计的关键思想,并通过修改其小碎片来优化分子。我们的模型了解如何识别待优化的碎片以及如何通过学习具有良好和不良性质的分子的差异来修改此类碎片。在优化新分子时,我们的模型将学习信号应用于在片段的预测位置解码优化的片段。我们还将多个这样的模型构造成管道,使得管道中的每个模型能够优化一个片段,因此整个流水线能够在需要时改变多个分子片段。我们将我们的模型与基准数据集的其他最先进的方法进行比较,并证明我们的方法在中等分子相似度约束下具有超过80%的性质改善,在高分子相似度约束下具有超过80%的财产改善。 。
translated by 谷歌翻译
它是科学技术的基础,能够预测化学反应及其性质。为实现此类技能,重要的是要培养良好的化学反应表示,或者可以自动从数据中学习此类表示的良好深度学习架构。目前没有普遍和广泛采用的方法,可强健地代表化学反应。大多数现有方法患有一个或多个缺点,例如:(1)缺乏普遍性; (2)缺乏稳健性; (3)缺乏可解释性;或(4)需要过度手动预处理。在这里,我们利用基于图的分子结构表示,以开发和测试一个超图注意神经网络方法,以一次解决反应表示和性能 - 预测问题,减轻了上述缺点。我们使用三个独立数据集化学反应评估三个实验中的这种超照片表示。在所有实验中,基于超图的方法与其他表示和它们相应的化学反应模型相匹配或优于相应的模型,同时产生可解释的多级表示。
translated by 谷歌翻译
阐明并准确预测分子的吸毒性和生物活性在药物设计和发现中起关键作用,并且仍然是一个开放的挑战。最近,图神经网络(GNN)在基于图的分子属性预测方面取得了显着进步。但是,当前基于图的深度学习方法忽略了分子的分层信息以及特征通道之间的关系。在这项研究中,我们提出了一个精心设计的分层信息图神经网络框架(称为hignn),用于通过利用分子图和化学合成的可见的无限元素片段来预测分子特性。此外,首先在Hignn体系结构中设计了一个插件功能的注意块,以适应消息传递阶段后自适应重新校准原子特征。广泛的实验表明,Hignn在许多具有挑战性的药物发现相关基准数据集上实现了最先进的预测性能。此外,我们设计了一种分子碎片的相似性机制,以全面研究Hignn模型在子图水平上的解释性,表明Hignn作为强大的深度学习工具可以帮助化学家和药剂师识别出设计更好分子的关键分子,以设计更好的分子,以设计出所需的更好分子。属性或功能。源代码可在https://github.com/idruglab/hignn上公开获得。
translated by 谷歌翻译
机器学习(ML)已经证明了用于准确和结晶材料的准确性能预测的承诺。为了化学结构的高度精确的ML型号的化学结构属性预测,需要具有足够样品的数据集。然而,获得昂贵的化学性质的获得和充分数据可以是昂贵的令人昂贵的,这大大限制了ML模型的性能。通过计算机视觉和黑暗语言处理中数据增强的成功,我们开发了奥古里希姆:数据八级化图书馆化学结构。引入了弃头晶系统和分子的增强方法,其可以对基于指纹的ML模型和图形神经网络(GNNS)进行脱颖而出。我们表明,使用我们的增强策略意义地提高了ML模型的性能,特别是在使用GNNS时,我们开发的增强件在训练期间可以用作广告插件模块,并在用不同的GNN实施时证明了有效性。模型通过Theauglichem图书馆。基于Python的封装我们实现了EugliChem:用于化学结构的数据增强库,可公开获取:https://github.com/baratilab/auglichem.1
translated by 谷歌翻译