Deep reinforcement learning for de novo drug design

We introduce an artificial intelligence approach to de novo design of molecules with desired physical or biological properties. We have devised and implemented a novel computational strategy for de novo design of molecules with desired properties termed ReLeaSE (Reinforcement Learning for Structural Evolution). On the basis of deep and reinforcement learning (RL) approaches, ReLeaSE integrates two deep neural networks—generative and predictive—that are trained separately but are used jointly to generate novel targeted chemical libraries. ReLeaSE uses simple representation of molecules by their simplified molecular-input line-entry system (SMILES) strings only. Generative models are trained with a stack-augmented memory network to produce chemically feasible SMILES strings, and predictive models are derived to forecast the desired properties of the de novo–generated compounds. In the first phase of the method, generative and predictive models are trained separately with a supervised learning algorithm. In the second phase, both models are trained jointly with the RL approach to bias the generation of new chemical structures toward those with the desired physical and/or biological properties. In the proof-of-concept study, we have used the ReLeaSE method to design chemical libraries with a bias toward structural complexity or toward compounds with maximal, minimal, or specific range of physical properties, such as melting point or hydrophobicity, or toward compounds with inhibitory activity against Janus protein kinase 2. The approach proposed herein can find a general use for generating targeted chemical libraries of novel compounds optimized for either a single desired property or multiple properties.

[1]  Richard E. Turner,et al.  Sequence Tutor: Conservative Fine-Tuning of Sequence Generation Models with KL-control , 2016, ICML.

[2]  Matthew E Welsch,et al.  Privileged scaffolds for library design and drug discovery. , 2010, Current opinion in chemical biology.

[3]  S. Pudenz,et al.  The Use of Hasse Diagrams as a Potential Approach for Inverse QSAR , 2001, SAR and QSAR in environmental research.

[4]  Juan Carlos Fernández,et al.  Multiobjective evolutionary algorithms to identify highly autocorrelated areas: the case of spatial distribution in financially compromised farms , 2014, Ann. Oper. Res..

[5]  Tristan Deleu,et al.  Learning Operations on a Stack with Neural Turing Machines , 2016, ArXiv.

[6]  Richard S. Sutton,et al.  Multi-step Reinforcement Learning: A Unifying Algorithm , 2017, AAAI.

[7]  J. Hendler,et al.  Science Magazine , 2009 .

[8]  James B. Morris Formal Languages and their Relation to Automata , 1970 .

[9]  Alán Aspuru-Guzik,et al.  Automatic Chemical Design Using a Data-Driven Continuous Representation of Molecules , 2016, ACS central science.

[10]  Alexandre Varnek,et al.  Estimation of the size of drug-like chemical space based on GDB-17 data , 2013, Journal of Computer-Aided Molecular Design.

[11]  Sergey Plis,et al.  Deep Learning Applications for Predicting Pharmacological Properties of Drugs and Drug Repurposing Using Transcriptomic Data. , 2016, Molecular pharmaceutics.

[12]  Mario Cazzola,et al.  A gain-of-function mutation of JAK2 in myeloproliferative disorders. , 2005, The New England journal of medicine.

[13]  Phil Blunsom,et al.  Learning to Transduce with Unbounded Memory , 2015, NIPS.

[14]  Ricardo Macarron,et al.  Critical review of the role of HTS in drug discovery. , 2006, Drug discovery today.

[15]  Petra Schneider,et al.  De Novo Design at the Edge of Chaos. , 2016, Journal of medicinal chemistry.

[16]  Ryan G. Coleman,et al.  ZINC: A Free Tool to Discover Chemistry for Biology , 2012, J. Chem. Inf. Model..

[17]  Thomas Blaschke,et al.  Molecular de-novo design through deep reinforcement learning , 2017, Journal of Cheminformatics.

[18]  Gisbert Schneider,et al.  Deep Learning in Drug Discovery , 2016, Molecular informatics.

[19]  David Ryan Koes,et al.  Protein-Ligand Scoring with Convolutional Neural Networks , 2016, Journal of chemical information and modeling.

[20]  Emilio Benfenati,et al.  SMILES in QSPR/QSAR Modeling: results and perspectives. , 2007, Current drug discovery technologies.

[21]  Maria F. Sassano,et al.  Automated design of ligands to polypharmacological profiles , 2012, Nature.

[22]  Emilio Benfenati,et al.  SMILES as an alternative to the graph in QSAR modelling of bee toxicity , 2007, Comput. Biol. Chem..

[23]  Alán Aspuru-Guzik,et al.  Optimizing distributions over molecular space. An Objective-Reinforced Generative Adversarial Network for Inverse-design Chemistry (ORGANIC) , 2017 .

[24]  Gisbert Schneider,et al.  Active-learning strategies in computer-assisted drug discovery. , 2015, Drug discovery today.

[25]  J. Reymond The chemical space project. , 2015, Accounts of chemical research.

[26]  C. Lipinski Lead- and drug-like compounds: the rule-of-five revolution. , 2004, Drug discovery today. Technologies.

[27]  Tony Y Zhang,et al.  Process chemistry: The science, business, logic, and logistics. , 2006, Chemical reviews.

[28]  Heinz-Ulrich Schmitt,et al.  Results and perspectives , 2005, Belligerent Reprisals.

[29]  Marwin H. S. Segler,et al.  Modelling Chemical Reasoning to Predict Reactions , 2016, Chemistry.

[30]  Geoffrey E. Hinton,et al.  Visualizing Data using t-SNE , 2008 .

[31]  J S Smith,et al.  ANI-1: an extensible neural network potential with DFT accuracy at force field computational cost , 2016, Chemical science.

[32]  Johann Gasteiger,et al.  A Graph-Based Genetic Algorithm and Its Application to the Multiobjective Evolution of Median Molecules , 2004, J. Chem. Inf. Model..

[33]  Allen D. Roses,et al.  Pharmacogenetics in drug discovery and development: a translational perspective , 2008, Nature Reviews Drug Discovery.

[34]  Thierry Kogej,et al.  Generating Focussed Molecule Libraries for Drug Discovery with Recurrent Neural Networks , 2017, ArXiv.

[35]  G. Bemis,et al.  The properties of known drugs. 1. Molecular frameworks. , 1996, Journal of medicinal chemistry.

[36]  Wolfgang Guba,et al.  Recent developments in de novo design and scaffold hopping. , 2008, Current opinion in drug discovery & development.

[37]  Kenta Hongo,et al.  Bayesian molecular design with a chemical language model , 2017, Journal of Computer-Aided Molecular Design.

[38]  Jason Yosinski,et al.  Deep neural networks are easily fooled: High confidence predictions for unrecognizable images , 2014, 2015 IEEE Conference on Computer Vision and Pattern Recognition (CVPR).

[39]  Peter Ertl,et al.  Estimation of synthetic accessibility score of drug-like molecules based on molecular complexity and fragment contributions , 2009, J. Cheminformatics.

[40]  George Papadatos,et al.  The ChEMBL bioactivity database: an update , 2013, Nucleic Acids Res..

[41]  R. M. Owen,et al.  An analysis of the attrition of drug candidates from four major pharmaceutical companies , 2015, Nature Reviews Drug Discovery.

[42]  Yanli Wang,et al.  PubChem BioAssay: 2017 update , 2016, Nucleic Acids Res..

[43]  Marina Krakovsky Reinforcement renaissance , 2016, Commun. ACM.

[44]  Vijay S. Pande,et al.  SIML: A Fast SIMD Algorithm for Calculating LINGO Chemical Similarities on GPUs and CPUs , 2010, J. Chem. Inf. Model..

[45]  Joan Bruna,et al.  Intriguing properties of neural networks , 2013, ICLR.

[46]  Michael S Lajiness,et al.  Assessment of the consistency of medicinal chemists in reviewing sets of compounds. , 2004, Journal of medicinal chemistry.

[47]  C. Krittanawong,et al.  Artificial Intelligence in Precision Cardiovascular Medicine. , 2017, Journal of the American College of Cardiology.

[48]  Igor V. Tetko,et al.  How Accurately Can We Predict the Melting Points of Drug-like Compounds? , 2014, J. Chem. Inf. Model..

[49]  Minoru Sakatsume,et al.  The Jak Kinases Differentially Associate with the and (Accessory Factor) Chains of the Interferon Receptor to Form a Functional Receptor Unit Capable of Activating STAT Transcription Factors (*) , 1995, The Journal of Biological Chemistry.

[50]  K. Johnson An Update. , 1984, Journal of food protection.

[51]  P Schneider,et al.  Multi-objective active machine learning rapidly improves structure–activity models and reveals new protein–protein interaction inhibitors† †Electronic supplementary information (ESI) available: Details about computational comparisons and all screening results. See DOI: 10.1039/c5sc04272k , 2016, Chemical science.

[52]  Noel M. O'Boyle Towards a Universal SMILES representation - A standard method to generate canonical SMILES based on the InChI , 2012, Journal of Cheminformatics.

[53]  Andrey Kazennov,et al.  The cornucopia of meaningful leads: Applying deep adversarial autoencoders for new molecule development in oncology , 2016, Oncotarget.

[54]  Geoffrey E. Hinton,et al.  Generating Text with Recurrent Neural Networks , 2011, ICML.

[55]  H. Jaap van den Herik,et al.  Games solved: Now and in the future , 2002, Artif. Intell..

[56]  Fei-Fei Li,et al.  Visualizing and Understanding Recurrent Networks , 2015, ArXiv.

[57]  Gisbert Schneider,et al.  Computer-based de novo design of drug-like molecules , 2005, Nature Reviews Drug Discovery.

[58]  Jeffrey Dean,et al.  Distributed Representations of Words and Phrases and their Compositionality , 2013, NIPS.

[59]  P. Dineen,et al.  Now and in the future. , 1970, AORN journal.

[60]  Sung Jin Cho,et al.  Rational Combinatorial Library Design. 2. Rational Design of Targeted Combinatorial Peptide Libraries Using Chemical Similarity Probe and the Inverse QSAR Approaches , 1998, J. Chem. Inf. Comput. Sci..

[61]  Jürgen Schmidhuber,et al.  Long Short-Term Memory , 1997, Neural Computation.

[62]  F. Lombardo,et al.  Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. , 2001, Advanced drug delivery reviews.

[63]  Pierre Baldi,et al.  Deep Architectures and Deep Learning in Chemoinformatics: The Prediction of Aqueous Solubility for Drug-Like Molecules , 2013, J. Chem. Inf. Model..

[64]  Tomas Mikolov,et al.  Inferring Algorithmic Patterns with Stack-Augmented Recurrent Nets , 2015, NIPS.

[65]  Ronald J. Williams,et al.  Simple Statistical Gradient-Following Algorithms for Connectionist Reinforcement Learning , 2004, Machine Learning.

[66]  Yoshua Bengio,et al.  Empirical Evaluation of Gated Recurrent Neural Networks on Sequence Modeling , 2014, ArXiv.

[67]  Demis Hassabis,et al.  Mastering the game of Go with deep neural networks and tree search , 2016, Nature.

[68]  E. Emanuel,et al.  The End of Radiology? Three Threats to the Future Practice of Radiology. , 2016, Journal of the American College of Radiology : JACR.

[69]  Vijay S. Pande,et al.  Low Data Drug Discovery with One-Shot Learning , 2016, ACS central science.

[70]  Alexandre Tkatchenko,et al.  Quantum-chemical insights from deep tensor neural networks , 2016, Nature Communications.

[71]  Thierry Kogej,et al.  Generating Focused Molecule Libraries for Drug Discovery with Recurrent Neural Networks , 2017, ACS central science.

[72]  Alexander Tropsha,et al.  Best Practices for QSAR Model Development, Validation, and Exploitation , 2010, Molecular informatics.

[73]  J. Dearden,et al.  QSAR modeling: where have you been? Where are you going to? , 2014, Journal of medicinal chemistry.

[74]  Alexander Tropsha,et al.  Trust, But Verify: On the Importance of Chemical Structure Curation in Cheminformatics and QSAR Modeling Research , 2010, J. Chem. Inf. Model..

[75]  Jonas Boström,et al.  Computational chemistry-driven decision making in lead generation. , 2006, Drug discovery today.

[76]  Hiromasa Kaneko,et al.  Inverse QSPR/QSAR Analysis for Chemical Structure Generation (from y to x) , 2016, J. Chem. Inf. Model..

[77]  A. Hopkins,et al.  Navigating chemical space for biology and medicine , 2004, Nature.