Multi-channel PINN: investigating scalable and transferable neural networks for drug discovery

Analysis of compound–protein interactions (CPIs) has become a crucial prerequisite for drug discovery and drug repositioning. In vitro experiments are commonly used in identifying CPIs, but it is not feasible to discover the molecular and proteomic space only through experimental approaches. Machine learning’s advances in predicting CPIs have made significant contributions to drug discovery. Deep neural networks (DNNs), which have recently been applied to predict CPIs, performed better than other shallow classifiers. However, such techniques commonly require a considerable volume of dense data for each training target. Although the number of publicly available CPI data has grown rapidly, public data is still sparse and has a large number of measurement errors. In this paper, we propose a novel method, Multi-channel PINN, to fully utilize sparse data in terms of representation learning. With representation learning, Multi-channel PINN can utilize three approaches of DNNs which are a classifier, a feature extractor, and an end-to-end learner. Multi-channel PINN can be fed with both low and high levels of representations and incorporates each of them by utilizing all approaches within a single model. To fully utilize sparse public data, we additionally explore the potential of transferring representations from training tasks to test tasks. As a proof of concept, Multi-channel PINN was evaluated on fifteen combinations of feature pairs to investigate how they affect the performance in terms of highest performance, initial performance, and convergence speed. The experimental results obtained indicate that the multi-channel models using protein features performed better than single-channel models or multi-channel models using compound features. Therefore, Multi-channel PINN can be advantageous when used with appropriate representations. Additionally, we pretrained models on a training task then finetuned them on a test task to figure out whether Multi-channel PINN can capture general representations for compounds and proteins. We found that there were significant differences in performance between pretrained models and non-pretrained models.

[1]  Mark McGann,et al.  FRED Pose Prediction and Virtual Screening Accuracy , 2011, J. Chem. Inf. Model..

[2]  Robert C. Glen,et al.  Classifying Molecules Using a Sparse Probabilistic Kernel Binary Classifier , 2011, J. Chem. Inf. Model..

[3]  Jorge Nocedal,et al.  On Large-Batch Training for Deep Learning: Generalization Gap and Sharp Minima , 2016, ICLR.

[4]  Wojciech Zaremba,et al.  An Empirical Exploration of Recurrent Network Architectures , 2015, ICML.

[5]  P. Bork,et al.  Drug Target Identification Using Side-Effect Similarity , 2008, Science.

[6]  Jürgen Schmidhuber,et al.  Framewise phoneme classification with bidirectional LSTM and other neural network architectures , 2005, Neural Networks.

[7]  Kei-Hoi Cheung,et al.  Bringing Web 2.0 to bioinformatics , 2008, Briefings Bioinform..

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

[9]  Jean-Louis Reymond,et al.  Enumeration of 166 Billion Organic Small Molecules in the Chemical Universe Database GDB-17 , 2012, J. Chem. Inf. Model..

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

[11]  Alexios Koutsoukas,et al.  Deep-learning: investigating deep neural networks hyper-parameters and comparison of performance to shallow methods for modeling bioactivity data , 2017, Journal of Cheminformatics.

[12]  María Martín,et al.  UniProt: A hub for protein information , 2015 .

[13]  Cathy H. Wu,et al.  UniProt: the Universal Protein knowledgebase , 2004, Nucleic Acids Res..

[14]  Fei Luo,et al.  Pairwise input neural network for target-ligand interaction prediction , 2014, 2014 IEEE International Conference on Bioinformatics and Biomedicine (BIBM).

[15]  Joseph Gomes,et al.  MoleculeNet: a benchmark for molecular machine learning† †Electronic supplementary information (ESI) available. See DOI: 10.1039/c7sc02664a , 2017, Chemical science.

[16]  Daniel R. Figueiredo,et al.  struc2vec: Learning Node Representations from Structural Identity , 2017, KDD.

[17]  Philip E. Bourne,et al.  Drug Discovery Using Chemical Systems Biology: Weak Inhibition of Multiple Kinases May Contribute to the Anti-Cancer Effect of Nelfinavir , 2011, PLoS Comput. Biol..

[18]  Heiga Zen,et al.  WaveNet: A Generative Model for Raw Audio , 2016, SSW.

[19]  Xiangrong Liu,et al.  Machine Learning for Drug-Target Interaction Prediction , 2018, Molecules.

[20]  Sepp Hochreiter,et al.  Fast and Accurate Deep Network Learning by Exponential Linear Units (ELUs) , 2015, ICLR.

[21]  Robert P. Sheridan,et al.  Deep Neural Nets as a Method for Quantitative Structure-Activity Relationships , 2015, J. Chem. Inf. Model..

[22]  W. P. Walters,et al.  Virtual Chemical Libraries. , 2018, Journal of medicinal chemistry.

[23]  Matthew P. Repasky,et al.  Glide: a new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. , 2004, Journal of medicinal chemistry.

[24]  Klaus-Robert Müller,et al.  Efficient BackProp , 2012, Neural Networks: Tricks of the Trade.

[25]  Gerard J. P. van Westen,et al.  Proteochemometric modeling as a tool to design selective compounds and for extrapolating to novel targets , 2011 .

[26]  Xiaomin Luo,et al.  TarFisDock: a web server for identifying drug targets with docking approach , 2006, Nucleic Acids Res..

[27]  Hao Ding,et al.  Similarity-based machine learning methods for predicting drug-target interactions: a brief review , 2014, Briefings Bioinform..

[28]  Xing Chen,et al.  In silico prediction of drug-target interaction networks based on drug chemical structure and protein sequences , 2017, Scientific Reports.

[29]  Geoffrey E. Hinton,et al.  Deep Learning , 2015, Nature.

[30]  George Papadatos,et al.  Activity, assay and target data curation and quality in the ChEMBL database , 2015, Journal of Computer-Aided Molecular Design.

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

[32]  Takaya Saito,et al.  The Precision-Recall Plot Is More Informative than the ROC Plot When Evaluating Binary Classifiers on Imbalanced Datasets , 2015, PloS one.

[33]  Hilde van der Togt,et al.  Publisher's Note , 2003, J. Netw. Comput. Appl..

[34]  John B. O. Mitchell,et al.  Predicting the mechanism of phospholipidosis , 2012, Journal of Cheminformatics.

[35]  Jimmy Ba,et al.  Adam: A Method for Stochastic Optimization , 2014, ICLR.

[36]  Sergey Ioffe,et al.  Rethinking the Inception Architecture for Computer Vision , 2015, 2016 IEEE Conference on Computer Vision and Pattern Recognition (CVPR).

[37]  Isidro Cortes-Ciriano,et al.  Polypharmacology modelling using proteochemometrics (PCM): recent methodological developments, applications to target families, and future prospects , 2015 .

[38]  Lin He,et al.  Exploring Off-Targets and Off-Systems for Adverse Drug Reactions via Chemical-Protein Interactome — Clozapine-Induced Agranulocytosis as a Case Study , 2011, PLoS Comput. Biol..

[39]  Andreas Bender,et al.  How Similar Are Similarity Searching Methods? A Principal Component Analysis of Molecular Descriptor Space , 2009, J. Chem. Inf. Model..

[40]  Mehmet Gönen,et al.  Predicting drug-target interactions from chemical and genomic kernels using Bayesian matrix factorization , 2012, Bioinform..

[41]  Andreas Bender,et al.  Recognizing Pitfalls in Virtual Screening: A Critical Review , 2012, J. Chem. Inf. Model..

[42]  David S. Goodsell,et al.  Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function , 1998, J. Comput. Chem..

[43]  Jure Leskovec,et al.  node2vec: Scalable Feature Learning for Networks , 2016, KDD.

[44]  Gerard J. P. van Westen,et al.  Benchmarking of protein descriptor sets in proteochemometric modeling (part 1): comparative study of 13 amino acid descriptor sets , 2013, Journal of Cheminformatics.

[45]  Andreas Verras,et al.  Is Multitask Deep Learning Practical for Pharma? , 2017, J. Chem. Inf. Model..

[46]  Lorenz C. Blum,et al.  Chemical space as a source for new drugs , 2010 .

[47]  Hege S. Beard,et al.  Glide: a new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screening. , 2004, Journal of medicinal chemistry.

[48]  George Papadatos,et al.  Beyond the hype: deep neural networks outperform established methods using a ChEMBL bioactivity benchmark set , 2017, bioRxiv.

[49]  Ehsaneddin Asgari,et al.  Continuous Distributed Representation of Biological Sequences for Deep Proteomics and Genomics , 2015, PloS one.

[50]  Jürgen Schmidhuber,et al.  LSTM: A Search Space Odyssey , 2015, IEEE Transactions on Neural Networks and Learning Systems.

[51]  Andreas Bender,et al.  Ligand-Target Prediction Using Winnow and Naive Bayesian Algorithms and the Implications of Overall Performance Statistics , 2008, J. Chem. Inf. Model..

[52]  Michael J. Keiser,et al.  Relating protein pharmacology by ligand chemistry , 2007, Nature Biotechnology.

[53]  Dong-Sheng Cao,et al.  Large-scale prediction of drug-target interactions using protein sequences and drug topological structures. , 2012, Analytica chimica acta.

[54]  Andreas Bender,et al.  From in silico target prediction to multi-target drug design: current databases, methods and applications. , 2011, Journal of proteomics.

[55]  Emilio Soria Olivas,et al.  Handbook of Research on Machine Learning Applications and Trends : Algorithms , Methods , and Techniques , 2009 .

[56]  Navdeep Jaitly,et al.  Multi-task Neural Networks for QSAR Predictions , 2014, ArXiv.

[57]  R. Glen,et al.  Molecular similarity: a key technique in molecular informatics. , 2004, Organic & biomolecular chemistry.

[58]  Volkan Atalay,et al.  Recent applications of deep learning and machine intelligence on in silico drug discovery: methods, tools and databases , 2018, Briefings Bioinform..

[59]  The Uniprot Consortium,et al.  UniProt: a hub for protein information , 2014, Nucleic Acids Res..

[60]  Nitesh V. Chawla,et al.  metapath2vec: Scalable Representation Learning for Heterogeneous Networks , 2017, KDD.

[61]  Karen Spärck Jones A statistical interpretation of term specificity and its application in retrieval , 2021, J. Documentation.

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

[63]  Sabrina Jaeger,et al.  Mol2vec: Unsupervised Machine Learning Approach with Chemical Intuition , 2018, J. Chem. Inf. Model..

[64]  Yoshihiro Yamanishi,et al.  Large-Scale Prediction of Beneficial Drug Combinations Using Drug Efficacy and Target Profiles , 2015, J. Chem. Inf. Model..

[65]  T. Lundstedt,et al.  Proteochemometrics modeling of the interaction of amine G-protein coupled receptors with a diverse set of ligands. , 2002, Molecular pharmacology.

[66]  Tianqi Chen,et al.  Empirical Evaluation of Rectified Activations in Convolutional Network , 2015, ArXiv.

[67]  Gang Fu,et al.  PubChem Substance and Compound databases , 2015, Nucleic Acids Res..

[68]  David D. Cox,et al.  On the information bottleneck theory of deep learning , 2018, ICLR.

[69]  Dingfeng Wu,et al.  The recent progress in proteochemometric modelling: focusing on target descriptors, cross‐term descriptors and application scope , 2017, Briefings Bioinform..

[70]  H. Yabuuchi,et al.  Analysis of multiple compound–protein interactions reveals novel bioactive molecules , 2011, Molecular systems biology.

[71]  Robert P. Sheridan,et al.  Random Forest: A Classification and Regression Tool for Compound Classification and QSAR Modeling , 2003, J. Chem. Inf. Comput. Sci..