Machine Learning Papers: Molecules

A self-attention based message passing neural network for predicting molecular lipophilicity and aqueous solubility - Tang et al 2020

Efficient and accurate prediction of molecular properties, such as lipophilicity and solubility, is highly desirable for rational compound design in chemical and pharmaceutical industries. To this end, we build and apply a graph-neural-network framework called self-attention-based message-passing neural network (SAMPN) to study the relationship between chemical properties and structures in an interpretable way. The main advantages of SAMPN are that it directly uses chemical graphs and breaks the black-box mold of many machine/deep learning methods. Specifically, its attention mechanism indicates the degree to which each atom of the molecule contributes to the property of interest, and these results are easily visualized. Further, SAMPN outperforms random forests and the deep learning framework MPN from Deepchem. In addition, another formulation of SAMPN (Multi-SAMPN) can simultaneously predict multiple chemical properties with higher accuracy and efficiency than other models that predict one specific chemical property. Moreover, SAMPN can generate chemically visible and interpretable results, which can help researchers discover new pharmaceuticals and materials. The source code of the SAMPN prediction pipeline is freely available at Github (https://github.com/tbwxmu/SAMPN).

DeepSMILES: An Adaptation of SMILES for Use in Machine-Learning of Chemical Structures - O'Boyle and Dalke 2018

Background
There has been increasing interest in the use of deep neural networks for de novo design of molecules with desired properties. A common approach is to train a generative model on SMILES strings and then use this to generate SMILES strings for molecules with a desired property. Unfortunately, these SMILES strings are often not syntactically valid due to elements of SMILES syntax that must occur in pairs.
 
Results
We describe a SMILES-like syntax called DeepSMILES that addresses two of the main reasons for invalid syntax when using a probabilistic model to generate SMILES strings. The DeepSMILES syntax avoids the problem of unbalanced parentheses by only using close parentheses, where the number of parentheses indicates the branch length. In addition, DeepSMILES avoids the problem of pairing ring closure symbols by using only a single symbol at the ring closing location, where the symbol indicates the ring size. We show that this syntax can be interconverted to/from SMILES with string processing without any loss of information, including stereo configuration.
 
Conclusion
We believe that DeepSMILES will be useful, not just for those using SMILES in deep neural networks, but also for other computational methods that use SMILES as the basis for generating molecular structures such as genetic algorithms.

A de novo molecular generation method using latent vector based generative adversarial network - Prykhodko et al. 2019

Deep learning methods applied to drug discovery have been used to generate novel structures. In this study, we propose a new deep learning architecture, LatentGAN, which combines an autoencoder and a generative adversarial neural network for de novo molecular design. We applied the method in two scenarios: one to generate random drug-like compounds and another to generate target-biased compounds. Our results show that the method works well in both cases. Sampled compounds from the trained model can largely occupy the same chemical space as the training set and also generate a substantial fraction of novel compounds. Moreover, the drug-likeness score of compounds sampled from LatentGAN is also similar to that of the training set. Lastly, generated compounds differ from those obtained with a Recurrent Neural Network-based generative model approach, indicating that both methods can be used complementarily.

MolGAN: An implicit generative model for small molecular graphs - De Cao and Kipf 2018

Deep generative models for graph-structured data offer a new angle on the problem of chemical synthesis: by optimizing differentiable models that directly generate molecular graphs, it is possible to side-step expensive search procedures in the discrete and vast space of chemical structures. We introduce MolGAN, an implicit, likelihood-free generative model for small molecular graphs that circumvents the need for expensive graph matching procedures or node ordering heuristics of previous likelihood-based methods. Our method adapts generative adversarial networks (GANs) to operate directly on graph-structured data. We combine our approach with a reinforcement learning objective to encourage the generation of molecules with specific desired chemical properties. In experiments on the QM9 chemical database, we demonstrate that our model is capable of generating close to 100% valid compounds. MolGAN compares favorably both to recent proposals that use string-based (SMILES) representations of molecules and to a likelihood-based method that directly generates graphs, albeit being susceptible to mode collapse.

Deep learning for molecular design - a review of the state of the art - Elton et al. 2019

In the space of only a few years, deep generative modeling has revolutionized how we think of artificial creativity, yielding autonomous systems which produce original images, music, and text. Inspired by these successes, researchers are now applying deep generative modeling techniques to the generation and optimization of molecules—in our review we found 45 papers on the subject published in the past two years. These works point to a future where such systems will be used to generate lead molecules, greatly reducing resources spent downstream synthesizing and characterizing bad leads in the lab. In this review we survey the increasingly complex landscape of models and representation schemes that have been proposed. The four classes of techniques we describe are recursive neural networks, autoencoders, generative adversarial networks, and reinforcement learning. After first discussing some of the mathematical fundamentals of each technique, we draw high level connections and comparisons with other techniques and expose the pros and cons of each. Several important high level themes emerge as a result of this work, including the shift away from the SMILES string representation of molecules towards more sophisticated representations such as graph grammars and 3D representations, the importance of reward function design, the need for better standards for benchmarking and testing, and the benefits of adversarial training and reinforcement learning over maximum likelihood based training.

DeepDTA: Deep Drug-Target Binding Affinity Prediction - Öztürk et al 2018

The identification of novel drug-target (DT) interactions is a substantial part of the drug discovery process. Most of the computational methods that have been proposed to predict DT interactions have focused on binary classification, where the goal is to determine whether a DT pair interacts or not. However, protein-ligand interactions assume a continuum of binding strength values, also called binding affinity and predicting this value still remains a challenge. The increase in the affinity data available in DT knowledge-bases allows the use of advanced learning techniques such as deep learning architectures in the prediction of binding affinities. In this study, we propose a deep-learning based model that uses only sequence information of both targets and drugs to predict DT interaction binding affinities. The few studies that focus on DT binding affinity prediction use either 3D structures of protein-ligand complexes or 2D features of compounds. One novel approach used in this work is the modeling of protein sequences and compound 1D representations with convolutional neural networks (CNNs). The results show that the proposed deep learning based model that uses the 1D representations of targets and drugs is an effective approach for drug target binding affinity prediction. The model in which high-level representations of a drug and a target are constructed via CNNs achieved the best Concordance Index (CI) performance in one of our larger benchmark data sets, outperforming the KronRLS algorithm and SimBoost, a state-of-the-art method for DT binding affinity prediction.

Prediction of drug–target binding affinity using graph neural networks - Nguyen et al 2019

Background
While the development of new drugs is costly, time consuming, and often accompanied with safety issues, drug repurposing, where old drugs with established safety are used for medical conditions other than originally developed, is an attractive alternative. Then, how the old drugs work on new targets becomes a crucial part of drug repurposing and gains much of interest. Several statistical and machine learning models have been proposed to estimate drug–target binding affinity and deep learning approaches have been shown to be among state-of-the-art methods. However, drugs and targets in these models have been commonly represented in 1D strings, regardless the fact that molecules are by nature formed by the chemical bonding of atoms.

Method
In this work, we propose GraphDTA to capture the structural information of drugs, possibly enhancing the predictive power of the affinity. In particular, unlike competing methods, drugs are represented as graphs and graph convolutional networks are used to learn drug–target binding affinity. We trial our method on two benchmark datasets of drug–target binding affinity and compare the performance with state-of-the-art models in the research field.

Convolutional neural network based on SMILES representation of compounds for detecting chemical motif - Hirohara et al 2018

Background
Previous studies have suggested deep learning to be a highly effective approach for screening lead compounds for new drugs. Several deep learning models have been developed by addressing the use of various kinds of fingerprints and graph convolution architectures. However, these methods are either advantageous or disadvantageous depending on whether they (1) can distinguish structural differences including chirality of compounds, and (2) can automatically discover effective features.

Results
We developed another deep learning model for compound classification. In this method, we constructed a distributed representation of compounds based on the SMILES notation, which linearly represents a compound structure, and applied the SMILES-based representation to a convolutional neural network (CNN). The use of SMILES allows us to process all types of compounds while incorporating a broad range of structure information, and representation learning by CNN automatically acquires a low-dimensional representation of input features. In a benchmark experiment using the TOX 21 dataset, our method outperformed conventional fingerprint methods, and performed comparably against the winning model of the TOX 21 Challenge. Multivariate analysis confirmed that the chemical space consisting of the features learned by SMILES-based representation learning adequately expressed a richer feature space that enabled the accurate discrimination of compounds. Using motif detection with the learned filters, not only important known structures (motifs) such as protein-binding sites but also structures of unknown functional groups were detected.

Quantum-Chemical Insights from Deep Tensor Neural Networks - Schutt et al 2016

Learning from data has led to paradigm shifts in a multitude of disciplines, including web, text, and image search, speech recognition, as well as bioinformatics. Can machine learning enable similar breakthroughs in understanding quantum many-body systems? Here we develop an efficient deep learning approach that enables spatially and chemically resolved insights into quantum-mechanical observables of molecular systems. We unify concepts from many-body Hamiltonians with purpose-designed deep tensor neural networks (DTNN), which leads to size-extensive and uniformly accurate (1 kcal/mol) predictions in compositional and configurational chemical space for molecules of intermediate size. As an example of chemical relevance, the DTNN model reveals a classification of aromatic rings with respect to their stability — a useful property that is not contained as such in the training dataset. Further applications of DTNN for predicting atomic energies and local chemical potentials in molecules, reliable isomer energies, and molecules with peculiar electronic structure demonstrate the high potential of machine learning for revealing novel insights into complex quantum-chemical systems.

Inverse molecular design using machine learning: Generative models for matter engineering - Sanchez-Lengeling and Aspuru-Guzik 2018

The discovery of new materials can bring enormous societal and technological progress. In this context, exploring completely the large space of potential materials is computationally intractable. Here, we review methods for achieving inverse design, which aims to discover tailored materials from the starting point of a particular desired functionality. Recent advances from the rapidly growing field of artificial intelligence, mostly from the subfield of machine learning, have resulted in a fertile exchange of ideas, where approaches to inverse molecular design are being proposed and employed at a rapid pace. Among these, deep generative models have been applied to numerous classes of materials: rational design of prospective drugs, synthetic routes to organic compounds, and optimization of photovoltaics and redox flow batteries, as well as a variety of other solid-state materials.

Automatic Chemical Design Using a Data-Driven Continuous Representation of Molecules - Gómez-Bombarelli et al 2017

We report a method to convert discrete representations of molecules to and from a multidimensional continuous representation. This model allows us to generate new molecules for efficient exploration and optimization through open-ended spaces of chemical compounds. A deep neural network was trained on hundreds of thousands of existing chemical structures to construct three coupled functions: an encoder, a decoder, and a predictor. The encoder converts the discrete representation of a molecule into a real-valued continuous vector, and the decoder converts these continuous vectors back to discrete molecular representations. The predictor estimates chemical properties from the latent continuous vector representation of the molecule. Continuous representations of molecules allow us to automatically generate novel chemical structures by performing simple operations in the latent space, such as decoding random vectors, perturbing known chemical structures, or interpolating between molecules. Continuous representations also allow the use of powerful gradient-based optimization to efficiently guide the search for optimized functional compounds. We demonstrate our method in the domain of drug-like molecules and also in a set of molecules with fewer that nine heavy atoms.

SMILES Enumeration as Data Augmentation for Neural Network Modeling of Molecules - Bjerrum

Simplified Molecular Input Line Entry System (SMILES) is a single line text representation of a unique molecule. One molecule can however have multiple SMILES strings, which is a reason that canonical SMILES have been defined, which ensures a one to one correspondence between SMILES string and molecule. Here the fact that multiple SMILES represent the same molecule is explored as a technique for data augmentation of a molecular QSAR dataset modeled by a long short term memory (LSTM) cell based neural network. The augmented dataset was 130 times bigger than the original. The network trained with the augmented dataset shows better performance on a test set when compared to a model built with only one canonical SMILES string per molecule. The correlation coefficient R2 on the test set was improved from 0.56 to 0.66 when using SMILES enumeration, and the root mean square error (RMS) likewise fell from 0.62 to 0.55. The technique also works in the prediction phase. By taking the average per molecule of the predictions for the enumerated SMILES a further improvement to a correlation coefficient of 0.68 and a RMS of 0.52 was found.

Machine learning for molecular and materials science - Butler et al

Here we summarize recent progress in machine learning for the chemical sciences. We outline machine-learning techniques that are suitable for addressing research questions in this domain, as well as future directions for the field. We envisage a future in which the design, synthesis, characterization and application of molecules and materials is accelerated by artificial intelligence.

Convolutional Networks on Graphs for Learning Molecular Fingerprints - Duvenaud et al

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.

Neural Message Passing for Quantum Chemistry - Gilmer et al

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.

FP2VEC: a new molecular featurizer for learning molecular properties Woosung Jeon, Dongsup Kim

One of the most successful methods for predicting the properties of chemical compounds is the quantitative structure–activity relationship (QSAR) methods. The prediction accuracy of QSAR models has recently been greatly improved by employing deep learning technology. Especially, newly developed molecular featurizers based on graph convolution operations on molecular graphs significantly outperform the conventional extended connectivity fingerprints (ECFP) feature in both classification and regression tasks, indicating that it is critical to develop more effective new featurizers to fully realize the power of deep learning techniques. Motivated by the fact that there is a clear analogy between chemical compounds and natural languages, this work develops a new molecular featurizer, FP2VEC, which represents a chemical compound as a set of trainable embedding vectors.

MoleculeNet: A Benchmark for Molecular Machine Learning - Wu et al 2018

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 open source library). MoleculeNet benchmarks demonstrate that learnable representations are powerful tools for molecular machine learning and broadly offer the best performance. However, this result comes with caveats. Learnable representations still struggle to deal with complex tasks under data scarcity and highly imbalanced classification. For quantum mechanical and biophysical datasets, the use of physics-aware featurizations can be more important than choice of particular learning algorithm.

Junction Tree Variational Autoencoder for Molecular Graph Generation - Jin et al

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.

druGAN: An Advanced Generative Adversarial Autoencoder Model for de Novo Generation of New Molecules with Desired Molecular Properties in Silico - Kadurin et al

Deep generative adversarial networks (GANs) are the emerging technology in drug discovery and biomarker development. In our recent work, we demonstrated a proof-of-concept of implementing deep generative adversarial autoencoder (AAE) to identify new molecular fingerprints with predefined anticancer properties. Another popular generative model is the variational autoencoder (VAE), which is based on deep neural architectures. In this work, we developed an advanced AAE model for molecular feature extraction problems, and demonstrated its advantages compared to VAE in terms of (a) adjustability in generating molecular fingerprints; (b) capacity of processing very large molecular data sets; and (c) efficiency in unsupervised pretraining for regression model. Our results suggest that the proposed AAE model significantly enhances the capacity and efficiency of development of the new molecules with specific anticancer properties using the deep generative models.

Chemception: A Deep Neural Network with Minimal Chemistry Knowledge Matches the Performance of Expert-developed QSAR/QSPR Models - Goh et al 2017

In the last few years, we have seen the transformative impact of deep learning in many applications, particularly in speech recognition and computer vision. Inspired by Google’s Inception-ResNet deep convolutional neural network (CNN) for image classification, we have developed “Chemception”, a deep CNN for the prediction of chemical properties, using just the images of 2D drawings of molecules. We develop Chemception without providing any additional explicit chemistry knowledge, such as basic concepts like periodicity, or advanced features like molecular descriptors and fingerprints. We then show how Chemception can serve as a general-purpose neural network architecture for predicting toxicity, activity, and solvation properties when trained on a modest database of 600 to 40,000 compounds. When compared to multi-layer perceptron (MLP) deep neural networks trained with ECFP fingerprints, Chemception slightly outperforms in activity and solvation prediction and slightly underperforms in toxicity prediction. Having matched the performance of expert-developed QSAR/QSPR deep learning models, our work demonstrates the plausibility of using deep neural networks to assist in computational chemistry research, where the feature engineering process is performed primarily by a deep learning algorithm.

How Much Chemistry Does a Deep Neural Network Need to Know to Make Accurate Predictions? - Goh et al 2018

The meteoric rise of deep learning models in computer vision research, having achieved human-level accuracy in image recognition tasks is firm evidence of the impact of representation learning of deep neural networks. In the chemistry domain, recent advances have also led to the development of similar CNN models, such as Chemception, that is trained to predict chemical properties using images of molecular drawings. In this work, we investigate the effects of systematically removing and adding localized domain-specific information to the image channels of the training data. By augmenting images with only 3 additional basic information, and without introducing any architectural changes, we demonstrate that an augmented Chemception (AugChemception) outperforms the original model in the prediction of toxicity, activity, and solvation free energy. Then, by altering the information content in the images, and examining the resulting model’s performance, we also identify two distinct learning patterns in predicting toxicity/activity as compared to solvation free energy. These patterns suggest that Chemception is learning about its tasks in the manner that is consistent with established knowledge. Thus, our work demonstrates that advanced chemical knowledge is not a pre-requisite for deep learning models to accurately predict complex chemical properties.

Deep Architectures and Deep Learning in Chemoinformatics: The Prediction of Aqueous Solubility for Drug-Like Molecules - Lusci at al 2013

Shallow machine learning methods have been applied to chemoinformatics problems with some success. As more data becomes available and more complex problems are tackled, deep machine learning methods may also become useful. Here, we present a brief overview of deep learning methods and show in particular how recursive neural network approaches can be applied to the problem of predicting molecular properties. However, molecules are typically described by undirected cyclic graphs, while recursive approaches typically use directed acyclic graphs. Thus, we develop methods to address this discrepancy, essentially by considering an ensemble of recursive neural networks associated with all possible vertex-centered acyclic orientations of the molecular graph. One advantage of this approach is that it relies only minimally on the identification of suitable molecular descriptors because suitable representations are learned automatically from the data. Several variants of this approach are applied to the problem of redicting aqueous solubility and tested on four benchmark data sets. Experimental results show that the performance of the deep learning methods matches or exceeds the performance of other state-of-the-art methods according to several evaluation metrics and expose the fundamental limitations arising from training sets that are too small or too noisy. A Web-based predictor, AquaSol, is available online through the ChemDB portal (cdb.ics.uci.edu) together with additional material.

Molecular Graph Convolutions: Moving Beyond Fingerprints - Kearnes et al 2016

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 data-driven 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.