key: cord-103528-3tib5o1m authors: Ahmed, Asad; Mam, Bhavika; Sowdhamini, Ramanathan title: DEELIG: A Deep Learning-based approach to predict protein-ligand binding affinity date: 2020-09-28 journal: bioRxiv DOI: 10.1101/2020.09.28.316224 sha: doc_id: 103528 cord_uid: 3tib5o1m Protein-ligand binding prediction has extensive biological significance. Binding affinity helps in understanding the degree of protein-ligand interactions and has wide protein applications. Protein-ligand docking using virtual screening and molecular dynamic simulations are required to predict the binding affinity of a ligand to its cognate receptor. In order to perform such analyses, it requires intense computational power and it becomes impossible to cover the entire chemical space of small molecules. It has been aided by a shift towards using Machine Learning-based methodologies that aids in binding prediction using regression. Recent developments using deep learning has enabled us to make sense of massive amounts of complex datasets. Herein, the ability of the model to “learn” intrinsic patterns in a complex plane of data is the strength of the approach. Here, we have incorporated Convolutional Neural Networks that find spatial relationships among data to help us predict affinity of binding of proteins in whole superfamilies towards a diverse set of ligands. The models were trained and validated using a detailed methodology for feature extraction. We have also tested DEELIG on protein complexes relevant to the current public health scenario. Our approach to network construction and training on protein-ligand dataset prepared in-house has provided significantly better results than previously existing methods in the field. Proteins are a diverse class of dynamic macromolecular structures in living organisms and are essential for the biochemistry and physiology of the organism. Depending on their functional role (s), proteins may bind to other proteins, peptides, nucleic acids and non-peptide ligands with varying affinities. Determining protein-ligand affinity helps in understanding the reaction mechanism and kinetics of the reaction and has applications in drug development and pharmacology (1) . Protein-ligand interaction is measured in terms of Binding affinity. The stronger the readout for binding affinity, the stronger the interaction between protein and ligand may be inferred. It is quantified in terms of Inhibition constant (Ki), dissociation constant (Kd), changes in free energy measures (delta G, delta H) and (IC50) (2) . Predicting binding affinity between a protein and ligand complements experimental approaches and is usually used as a start-point for the latter. Prediction-based approaches are also useful in cases where experimental determination of binding affinity may not be feasible. Classical prediction methods to score free binding energies of small ligands to biological macromolecules such as MM/GBSA and MM/PBSA typically rely on molecular dynamic simulations for calculations and aid in silico docking and virtual screening as well as experimental approaches. However, there is a trade-off between computational resources and accuracy (3) . With a recent shift towards the use of machine learning and deep-learning based methods in the field of structural biology, making biologically significant predictions using regression and 'learning' intrinsic patterns in a complex plane of available data has led to resourceoptimal predictions without compromising on accuracy. Deep learning has been known to learn representations and patterns in complex data forms. Our aim was to apply deep learning to predict binding affinity of protein-ligand interaction. Convolutional Neural Networks (CNN) are deep neural networks that use an input layer, output later as well as convolutional hidden layer(s). The first CNN was incorporated by LeCunn in 1998 (4) the connectivity pattern of which was inspired by the elegant experiments of Hubert and Weisel on the mammalian visual cortex in the 1960s (5) . With the growing technical advancements and massive amounts of data, CNNs have emerged popular in biological fields in the recent decade with various applications (6) . In our study, we have used CNNs to provide a quantitative estimate of protein-ligand binding using various sets of features corresponding to protein and ligand respectively by finding spatial relationships amongst the data. Our approach was validated using ligand-bound complexes from kinases superfamily in the PDB. Kinases belong to a class of enzymes required for substrate dependent phosphorylation. They are represented across diverse cellular functions like signaling, differentiation, glycolysis (7) . We have also tested our model on COVID-19 main protease (8) of the novel coronavirus strain complexed with various inhibitors of which binding affinities have not been predicted or experimentally determined so far. The raw data for our novel database was obtained from RCSB PDB (9) database, where following were selected as the query parameters. These criteria resulted in a list of 5464 protein PDB IDs, 2568 complexed ligand (s) and corresponding binding affinity values. The search results include the structures present in PDBdatabase, PDBBind (10, 11, 12) , PDBMoad (13, 14) and scPDB (15) for its results. Initial raw data database created contained protein structures in PDB format, protein sequences in FASTA format, ligand in SDF format and binding affinity values of corresponding protein-ligand pairs for 5464 complexes. The PDB, FASTA and SDF files filtered were further processed to refine our novel dataset, as shown in Figure 1 . Protein-ligand complexes were 5,464 in number and corresponded to 29,650 complex unique chain-ligand pairs. Binding affinity values were obtained from the RCSB database and protein chain-ligand pairs with corresponding binding affinity as 0 were discarded to reduce statistical errors. This narrowed down the total complexes to 4,750 protein-ligand pairs. Pocket information was extracted from the protein using Ghecom (16) and converted to MOL2 format using Chimera (17) , which narrowed our results to 4699 pocket-ligand pairs. It narrowed down the size of the dataset to 4286 pocket-ligand pairs. We discarded other protein-ligand pairs with missing PSSM profiles, secondary structure or dihedral angle information. It resulted in a total of 4041 pocket-ligand pairs, which corresponds to 7414 pocket-ligand pairs containing unique chains. Training the deep learning network on raw information is known to result in longer time for convergence and less accuracy. We followed a conventional methodology for feature extraction and used the deep learning framework to learn the interaction between the proteinpocket and ligand for their affinity prediction. A comprehensive two-level feature extraction methodology, one at the atomic level and the other at the level of amino acids utilizing structural information and protein sequence respectively. • 9 Bit 1 hot or all null hot encoding for atom types: B, C, N, O, P, S, Se, halogen and metal. • 1 integer for hybridization • 1 integer representing the number of bonds with heavy atoms • 1 integer representing the number of bonds with hetero atoms • 5 bits (1 if present) encoding properties defined with SMARTS patterns: hydrophobic, aromatic, acceptor, donor and ring • 1 float for partial charges • 1 integer to distinguish between ligand as -1 and protein as 1 We utilized the sequence information of protein to get more features about the protein pocket-ligand interaction. • Position-Specific Scoring Matrix (PSSM): PSSM is a matrix that represents the probability of mutation at each point of the sequence. It gives a 20 bit-probability for each amino acid at each location. PSSM profiles were obtained using PSI-BLAST (18) with SwissProt as subject database and E-value threshold as 0.001. Chains with less than 50 amino acids were removed from the input dataset. • Relative Solvent Accessibility (RSA): It is encoded by 1 bit of information for each amino acid that provides whether it is buried or exposed to the solvent. We set a threshold of 25% in RSA values. RSA was obtained using NACCESS (19) . • Secondary Structure: It is encoded by 1 bit of information about the structure as coil, helix or plate and was predicted using the DSSP (20, 21). • Dihedral Angles: It is encoded by 2 bits of information with phi / psi angles of each of the amino acids and was predicted using DSSP (20, 21) for obtaining dihedral angles. Standard ligand features were calculated for ligands in our dataset using PADEL (22) and 1D, 2D and chemical fingerprints, which includes hybridisation, atom pair interaction, counts of various functional group. We also used QikProp (34) and CANVAS (23) to derive ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) properties, which includes the physical properties, solubility and partition coefficients. The exhaustive list of every property calculated is given in the appendix. It results in a 1D array of 14,716 dimensions containing the various properties of a given ligand. This is used as a feature vector representing the ligand represented in MOL2 format. The three-dimensional co-ordinates of atoms were converted into a 3D grid of resolution 10Å with 1Å spacing between the two axes centered along the centroid of the ligand. Atoms outside each such grid were discarded. The atoms lying inside the grid were rounded up to the nearest coordinate of the grid where features of corresponding atoms that lay in the same coordinates were added up. This resulted in projecting ligand-interacting residues into a three-dimensional cube with features representing the atomic as well as protein-based properties of each atom of the protein pocket. Features were calculated at the atomic level (Section 4.1.1) corresponding to each atom of an amino acid and ligand. A 19-bit vector was calculated that uniquely identified each of the atoms in the 3D co-ordinates of a given protein-pocket and ligand complex. A 4D tensor each of size m x m x m x 19, i.e. the 3 coordinates (x, y, z) and the features, where m represents the number of atoms present in a complex was constructed as the feature vector representing the given protein pocket-ligand. The 4D vector contains the protein-pocket features and was converted to a 3D grid using grid featurization (Section 4.3). The 3D-featurized grid is essentially a 4D tensor, where the coordinates are approximated to the points on the grid. The dataset is converted to vectors and is divided into training:validation:test sets in ratio 80:10:10. Convolutional Neural Networks (24) have been used to capture spatial features in an image. We use CNNs to capture the interaction between ligand and protein atoms in threedimensional space. A network was constructed ( Figure 2 ) with a 3D CNN of varying channel sizes of [64, 128, 256] with non-linear activation ReLU after each layer, each 3D CNN had a filter of 5Å cube which was used to perform convolution operations. MaxPool (25) layer that acts in three dimensions to lower the dimension with a pool size of 2Å cube and Batch Normalization (26) layer is added after each CNN layer, this in turn decreases the training time and helps in faster convergence. The latent features learnt from the above CNN layers were then flattened and used for calculating the binding affinity of the protein pocket-ligand pair. The CNN derives the relation among the 3D coordinates and their features, which would correspond well to the binding affinities of complexes. The features from the last CNN layer are then flattened out, and passed through a fully connected neural network having the number of neurons as [1000, 500, 250] with ReLU as non-linearity after each layer. Dropout (25) is added after each layer to prevent overfitting by forcing the neural network to learn various other pathways by randomly assigning neurons to zero, 0.50 as Dropout threshold. Dense network predicts a regressive value of Binding Affinity, corresponding to a single neuron output. Training framework is shown in Figure 2 and a detailed layer network is shown in Figure 4 (a). The featurized protein-pocket grid formed was rotated to all 24 combinations possible, such that the network is able learn in an orientation invariant form. The network was trained by taking Mean Square error between the predicted and actual values as a loss function. The network was optimized using Adam (27) as the optimizer with a learning rate of 1e-5 and weight decay of 0.001 for 20 epochs. Network was trained on an Nvidia Pascal GPU using Pytorch (28) as the framework. Features were calculated at the amino acid level (Section 4.1.2) and were concatenated alongside the atomic level features (Section 4.1.1) to each atom of amino acid. It results in a 44-bit vector uniquely identifying each of the atoms in the 3D co-ordinates of a given protein. A 4D tensor each of sizes m x m x m x 44, i.e. the 3 coordinates (x, y, z) and the features, where m represents the number of atoms present in a complex is constructed as the feature vector of protein pocket. The 4D vector contains the protein-pocket features, it was converted to a 3D grid using grid featurization (Section 4.3). The 3D featurized grid is essentially a 4D tensor, where the coordinates are approximated to the points on the grid. The ligands were separately featurized by calculating the ligand properties (Section 4.2), which results in a 1D tensor. The dataset is converted to vectors and is divided into training:validation:test sets in ratio 80:10:10. A multi-input network was constructed (29) Training framework is shown in Figure 3 and a detailed layer network is shown in Figure 4 (b) The featurized protein-pocket grid formed was rotated to all 24 combinations possible, such that the network is able to learn in an orientation invariant form. The featurized protein pocket-ligand pair of training set was passed through corresponding the network and trained by taking Mean Square error between the predicted and actual values as a loss function. The network was optimized using Adam (27) as the optimizer with a learning rate of 1e-5 and weight decay of 0.001. The network was trained on an Nvidia Pascal GPU using Pytorch (28) as the framework. The performance of the models was quantified using Mean Absolute error (MAE) and Root mean square error (RMSE). It was tested on validation and testing sets which were initially divided from our dataset as mentioned in the training section. Lower error corresponds to better learning capacity of the model. Standard deviation among the real and predicted values was also calculated. The MAE, RMSE and SD values are shown in Table 1 . For the purpose of training and testing models, one NVIDIA Tesla P100 GPU cluster was used. Computational time taken for featurization of dataset, training and testing were 52 hours, 22 hours and 8 minutes respectively. Two modules were trained. The first module was trained using a small set of features for protein and ligand, which were represented together in a 3D grid space. This approach has also been part of a previous study (29) . However, the previous study uses a restricted ligand set that does not involve larger ligands. Here we have used a diverse set of ligands as one of our inputs. With training of Atomic Model for 35 epochs, MAE score of 2.84 was achieved (Table 1) . We constructed another module that enabled us to improve on the ligand and protein based information. To this purpose, we used an increased feature vector size which amounted to 14716 bits in size for ligand and 44 bits for each atom of protein. With training of Composite Model for only 4 epochs, MAE score of 2.27 was achieved ( Table 1) . The performance of our model was further evaluated using ligand-bound complexes from the kinase superfamily from PDB. The composite model outperformed the atomic model significantly and with lower standard deviation. In light of the ongoing coronavirus pandemic, we tested protein-ligand complexes from the coronavirus (CoV) family. The COVID-19 main protease is a key enzyme for the novel strain of coronavirus that is being implicated in the pandemic. A recent study involved testing of invitro binding efficacy of coronavirus COVID-19 virus main protease (Mpro) with a potent reversible synthetic inhibitor, N3 (31) . However, the highly potent inhibition by N3 rendered the experimental determination of binding affinity not achievable. Using the structure of Mpro at high resolution (7BQY: 1.7 Angstrom), we have been able to predict the binding affinity of N3 to 3.1e+4 nanomolar (Table 3) . This value agrees with the observed high affinity in the course of recent experiments (31) . Another study has deposited the complex of the COVID-19 main protease with a broadspectrum inhibitor X77 (N-(4-tert-butylphenyl)-N-[ (1R)-2-(cyclohexylamino)-2-oxo-1-(pyridin-3-yl)ethyl]-1H-imidazole-4-carboxamide) (2020; unpublished). We used these complexes to predict their respective binding affinities as they have not been made available. Based on our model-based predictions, broad spectrum inhibitor X77 scores for highest affinity followed by ligands Z45617795, N3, Z31792168, Z1220452176 and Z219104216 in the order of decreasing binding affinity ( We propose a deep-learning based approach to predict ligand (eg., drug)-target binding affinity using only structures of target protein (PDB format) and ligand (SDF format) as inputs. Convolutional Neural Networks (CNN) were used to learn representations from the features extracted from these inputs and hidden layers in the affinity prediction task. We used two approaches to feature extraction-atomic level as well as composite level and compared their performance using the same network. Deep-learning based approaches have been implemented for prediction of binding affinity. One of the studies used atomic level features of complex in a CNN based framework for binding affinity prediction (32), while another study used protein sequence level features in a CNN based framework for prediction (33) . Another approach used as been to use feature learning along with gradient boosting algorithms to predict binding affinity (34) . Here, we provide a composite model that incorporates tripartite structural, sequence and atomic level features with those of the atomic and other chemical features of the ligand to predict binding affinity of a putative complex. We have trained two models to predict the binding affinity between protein and ligand in a given complex. This would help existing databases like RSCB PDB, PDBBind in filling missing binding affinity data for complexes. We have constructed a novel dataset that represents a diverse set of ligands and using a novel deep learning based approach we have achieved significant improvement in prediction of binding affinity of protein-ligand complexes. Interestingly, our approach performed better without ligand coordinates as input. To counter filtering or noise reduction in our dataset, our dataset constructed is smaller than PDBBind (35) but we have overcome the constraints on ligand selection part of a previous study (29) . Although our dataset contains 5464 complexes compared to 16,151 complexes found in PDBBind, the ligands used as part of our training include 452 unique ligands absent in PDBBind. This helps in achieving ligand diversity during training the CNN model. The similarity matrix constructed from the binary fingerprints of ligands used in the dataset supports our claim of improved ligand diversity in our dataset (Supplementary File S1). We have also eliminated the need of providing ligands in a complex form with protein. Thus a given protein pocket may be tested for the degree of binding for any given ligand. This can be extended to predicting potential binding partners for proteins in other superfamilies as well. It is also important to consider that docking score and pose is not a reliable correlation with MM/GBSA poses (36). DEELIG can be used for a member of any protein superfamily and a non-peptide ligand, the docking pose of which may or may not be known. The code repository for the project is publicly available at : https://github.com/asadahmedtech/DEELIG Binding affinity predictions through DEELIG can be extended to protein-ligand complexes of protein superfamilies where the affinity is quantitatively unknown due to experimental limitations or where the potential for binding is yet to be explored in vitro. A webserver to implement DEELIG for easy online access would be useful for the general scientific community and this will also be in the pipeline. A later version of DEELIG which is trained on peptide ligand dataset will also be worked on. Following properties of ligand were calculated using PADEL (22) • IP (ev) ( ionization potential) • EA (eV) (electron affinity) • #metab (likely metabolic reactions) • PSA (van der waals SA of polar N and O atoms) • #NandO, #ringatoms (number of atoms in rings) • #in34 (number of atoms in 3 or 4 membered rings) • #in56 (number of atoms in 5 or 6 membered rings) • #noncon (ring atoms cannot form conjugated aromatic bonds) • #nonHatm (heavy atoms-nonhydrogen atoms) • RuleOfThree • RuleOfFive (lipinski violations) • QPlogKhsa (binding to human serum albumin) • PercentHuman-OralAbsorption Recent improvements to binding moad: a resource for protein ligand binding affinities and structures Gapped blast and psi-blast: a new generation of protein database search programs Analysis and comparison of 2d fingerprints: insights into database screening performance using eight fingerprint methods The mm/pbsa and mm/gbsa methods to estimate ligandbinding affinities Expert opinion on drug discovery Simboost: a read-across approach for predicting drug-target binding affinities using gradient boosting machines Receptive fields, binocular interaction and functional architecture in the cat's visual cortex Batch normalization: Accelerating deep network training by reducing internal covariate shift Structure of M pro from SARS-CoV-2 and discovery of its inhibitors A series of pdb related databases for everyday needs sc-pdb: a 3d-database of ligandable binding sites-10 years on Dictionary of protein secondary structure: pattern recognition of hydrogen-bonded and geometrical features On the binding affinity of macromolecular interactions: daring to ask why proteins interact Detection of multiscale pockets on protein surfaces using mathematical morphology Adam: A method for stochastic optimization Imagenet classification with deep convolutional neural networks Binding moad (mother of all databases Gradient-based learning applied to document recognition Deepatom: A framework for protein-ligand binding affinity prediction Deep learning in bioinformatics Bindingmoad, a high-quality protein-ligand database Deepdta: deep drug-target binding affinity prediction Pytorch: Tensors and dynamic neural networks in python with strong gpu acceleration. PyTorch: Tensors and dynamic neural networks in Python with strong GPU acceleration Ucsf chimera-a visualization system for exploratory research and analysis The rcsb protein data bank: redesigned website and web services Very deep convolutional networks for largescale image recognition Development and evaluation of a deep learning model for protein-ligand binding affinity prediction The pdbbind database: collection of binding affinities for protein-ligand complexes with known three-dimensional structures The pdbbind database: methodologies and updates The pdbbind database: methodologies and updates Padel-descriptor: An open source software to calculate molecular descriptors and fingerprints Pdb-wide collection of binding data: current status of the pdbbind database Binding of nicotinoids and the related compounds to the insect nicotinic acetyicholine receptor Schrödinger Release 2020-3: QikProp, Schrödinger, LLC Supplementary Files a. Similarity matrix of ligands used in dataset (Supplementary File S1) b. Dataset details (Supplementary File S2) c. Dataset distribution AA acknowledges funding awarded by the Indian Academy of Sciences, Bangalore (2019).BM would like to acknowledge Tata Trusts-TDU Fellowship for PhD awarded to her from 2017 to 2019. All authors acknowledge NCBS for infrastructural support. The authors declare no conflict of interest.