Antonio de la Vega de León, Jürgen Bajorath

Similar documents
Antonio de la Vega de León, Jürgen Bajorath

Analysis of Activity Landscapes, Activity Cliffs, and Selectivity Cliffs. Jürgen Bajorath Life Science Informatics University of Bonn

Design and characterization of chemical space networks

Machine Learning Concepts in Chemoinformatics

QSAR Modeling of ErbB1 Inhibitors Using Genetic Algorithm-Based Regression

The Molecule Cloud - compact visualization of large collections of molecules

Condensed Graph of Reaction: considering a chemical reaction as one single pseudo molecule

Introduction. OntoChem

Emerging patterns mining and automated detection of contrasting chemical features

Analysis of a Large Structure/Biological Activity. Data Set Using Recursive Partitioning and. Simulated Annealing

An Integrated Approach to in-silico

has its own advantages and drawbacks, depending on the questions facing the drug discovery.

UniStra activities within the BigChem project:

Molecular Complexity Effects and Fingerprint-Based Similarity Search Strategies

Ignasi Belda, PhD CEO. HPC Advisory Council Spain Conference 2015

Similarity Search. Uwe Koch

Drug Design 2. Oliver Kohlbacher. Winter 2009/ QSAR Part 4: Selected Chapters

Analysis of Random Fragment Profiles for the Detection of Structure-Activity Relationships

An Enhancement of Bayesian Inference Network for Ligand-Based Virtual Screening using Features Selection

Building blocks for automated elucidation of metabolites: Machine learning methods for NMR prediction

Development of Pharmacophore Model for Indeno[1,2-b]indoles as Human Protein Kinase CK2 Inhibitors and Database Mining

Notes of Dr. Anil Mishra at 1

In Silico Investigation of Off-Target Effects

Chemical Data Retrieval and Management

A Tiered Screen Protocol for the Discovery of Structurally Diverse HIV Integrase Inhibitors

User Guide for LeDock

Improved Machine Learning Models for Predicting Selective Compounds

Data Quality Issues That Can Impact Drug Discovery

A Deep Convolutional Neural Network for Bioactivity Prediction in Structure-based Drug Discovery

Edges of Saturn s rings are fractal

Chemical Space. Space, Diversity, and Synthesis. Jeremy Henle, 4/23/2013

Evaluation of Molecular Similarity and Molecular Diversity Methods Using Biological Activity Data

Visualizing Correlation Networks

Computational chemical biology to address non-traditional drug targets. John Karanicolas

Exploring the black box: structural and functional interpretation of QSAR models.

CSD. Unlock value from crystal structure information in the CSD

CSD. CSD-Enterprise. Access the CSD and ALL CCDC application software

Distance-based test for uncertainty hypothesis testing

COMPARISON OF SIMILARITY METHOD TO IMPROVE RETRIEVAL PERFORMANCE FOR CHEMICAL DATA

Use of data mining and chemoinformatics in the identification and optimization of high-throughput screening hits for NTDs

Keywords: anti-coagulants, factor Xa, QSAR, Thrombosis. Introduction

Rapid Application Development using InforSense Open Workflow and Daylight Technologies Deliver Discovery Value

Navigating between patents, papers, abstracts and databases using public sources and tools

Chemical Space: Modeling Exploration & Understanding

Bridging the Dimensions:

Reaxys Medicinal Chemistry Fact Sheet

Navigation in Chemical Space Towards Biological Activity. Peter Ertl Novartis Institutes for BioMedical Research Basel, Switzerland

ENERGY MINIMIZATION AND CONFORMATION SEARCH ANALYSIS OF TYPE-2 ANTI-DIABETES DRUGS

CHAPTER 6 QUANTITATIVE STRUCTURE ACTIVITY RELATIONSHIP (QSAR) ANALYSIS

Research Article. Chemical compound classification based on improved Max-Min kernel

Analyzing Molecular Conformations Using the Cambridge Structural Database. Jason Cole Cambridge Crystallographic Data Centre

arxiv: v1 [cs.ds] 25 Jan 2016

Applying Bioisosteric Transformations to Predict Novel, High Quality Compounds

DivCalc: A Utility for Diversity Analysis and Compound Sampling

KNIME-based scoring functions in Muse 3.0. KNIME User Group Meeting 2013 Fabian Bös

Fast Hierarchical Clustering from the Baire Distance

Medicinal Chemist s Relationship with Additivity: Are we Taking the Fundamentals for Granted?

Follow links Class Use and other Permissions. For more information, send to:

Statistical concepts in QSAR.

Universities of Leeds, Sheffield and York

Visualization and manipulation of Matched Molecular Series for decision support

Selecting Diversified Compounds to Build a Tangible Library for Biological and Biochemical Assays

Chemical library design

QSAR/QSPR modeling. Quantitative Structure-Activity Relationships Quantitative Structure-Property-Relationships

Data Mining in the Chemical Industry. Overview of presentation

Molecular descriptors and chemometrics: a powerful combined tool for pharmaceutical, toxicological and environmental problems.

Development of a Structure Generator to Explore Target Areas on Chemical Space

Contents 1 Open-Source Tools, Techniques, and Data in Chemoinformatics

AMRI COMPOUND LIBRARY CONSORTIUM: A NOVEL WAY TO FILL YOUR DRUG PIPELINE

October 6 University Faculty of pharmacy Computer Aided Drug Design Unit

Large scale classification of chemical reactions from patent data

Trace inequalities for positive semidefinite matrices with centrosymmetric structure

The Case for Use Cases

Using Phase for Pharmacophore Modelling. 5th European Life Science Bootcamp March, 2017

Xia Ning,*, Huzefa Rangwala, and George Karypis

Chemoinformatics and information management. Peter Willett, University of Sheffield, UK

Introduction. Marcel Radermacher Algorithmen zur Visualisierung von Graphen

Master de Chimie M1S1 Examen d analyse statistique des données

Dr. Sander B. Nabuurs. Computational Drug Discovery group Center for Molecular and Biomolecular Informatics Radboud University Medical Centre

The PhilOEsophy. There are only two fundamental molecular descriptors

HSQC spectral based similarity matching of compounds using nearest neighbours and a fast discrete genetic algorithm

Using Self-Organizing maps to accelerate similarity search

Algorithms for Graph Visualization Force-Directed Algorithms

Kernel-based Machine Learning for Virtual Screening

Reaxys Pipeline Pilot Components Installation and User Guide

On modeling two immune effectors two strain antigen interaction

FCModeler: Dynamic Graph Display and Fuzzy Modeling of Regulatory and Metabolic Maps

Early Stages of Drug Discovery in the Pharmaceutical Industry

Virtual Libraries and Virtual Screening in Drug Discovery Processes using KNIME

Virtual affinity fingerprints in drug discovery: The Drug Profile Matching method

Fast similarity searching making the virtual real. Stephen Pickett, GSK

Analysis and visualization of protein-protein interactions. Olga Vitek Assistant Professor Statistics and Computer Science

Design and Synthesis of the Comprehensive Fragment Library

Hiromi Nishida. 1. Introduction. 2. Materials and Methods

Exploring the chemical space of screening results

Chemically Intelligent Experiment Data Management

Chemists are from Mars, Biologists from Venus. Originally published 7th November 2006

A powerful site for all chemists CHOICE CRC Handbook of Chemistry and Physics

HYPER ZAGREB INDEX OF BRIDGE AND CHAIN GRAPHS

EMPIRICAL VS. RATIONAL METHODS OF DISCOVERING NEW DRUGS

Transcription:

METHOD ARTICLE Design of chemical space networks incorporating compound distance relationships [version 2; referees: 2 approved] Antonio de la Vega de León, Jürgen Bajorath Department of Life Science Informatics, Rheinische Friedrich-Wilhelms-Universität, Bonn, Germany v2 First published: 04 Nov 2016, 5(Chem Inf Sci):2634 (doi: 10.12688/f1000research.10021.1) Latest published: 30 Nov 2016, 5(Chem Inf Sci):2634 (doi: 10.12688/f1000research.10021.2) Abstract Networks, in which nodes represent compounds and edges pairwise similarity relationships, are used as coordinate-free representations of chemical space. So-called chemical space networks (CSNs) provide intuitive access to structural relationships within compound data sets and can be annotated with activity information. However, in such similarity-based networks, distances between compounds are typically determined for layout purposes and clarity and have no chemical meaning. By contrast, inter-compound distances as a measure of dissimilarity can be directly obtained from coordinate-based representations of chemical space. Herein, we introduce a CSN variant that incorporates compound distance relationships and thus further increases the information content of compound networks. The design was facilitated by adapting the Kamada-Kawai algorithm. Kamada-Kawai networks are the first CSNs that are based on numerical similarity measures, but do not depend on chosen similarity threshold values. Keywords Chemical space, bioactive compounds, coordinate-free space representations, chemical similarity, similarity-based compound networks, force-directed network layouts This article is included in the Chemical Information Science gateway. Open Peer Review Referee Status: version 2 published 30 Nov 2016 version 1 published 04 Nov 2016 1 2 Discuss this article Comments (0) Invited Referees 1 2 report Alexandre Varnek, University of Strasbourg (UDS), France Gerhard Hessler, Sanofi-Aventis Deutschland GmbH., Germany report Corresponding author: Jürgen Bajorath ( bajorath@bit.uni-bonn.de) Competing interests: No competing interests were disclosed. Grant information: The author(s) declared that no grants were involved in supporting this work. Copyright: 2016 de la Vega de León A and Bajorath J. This is an open access article distributed under the terms of the Creative Commons Attribution Licence, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. How to cite this article: de la Vega de León A and Bajorath J. Design of chemical space networks incorporating compound distance relationships [version 2; referees: 2 approved] F1000Research 2016, 5(Chem Inf Sci):2634 (doi: 10.12688/f1000research.10021.2) First published: 04 Nov 2016, 5(Chem Inf Sci):2634 (doi: 10.12688/f1000research.10021.1) Page 1 of 10

REVISED Amendments from Version 1 We thank both reviewers for their comments. As suggested by Gerhard Hessler, we have added target names to the main text and further extended the discussion of threshold values and edge density variation for visualization and SAR analysis. See referee reports Introduction In chemoinformatics, molecular network representations have thus far mostly been applied to study similarity relationships between compounds and visualize structure-activity relationships (SARs) 1 3. In such networks, molecules are represented as nodes and edges indicate pairwise similarity relationships. Potency information can be added, for example, through node coloring, which provides a basis for SAR visualization 2. A prototypic network representation specifically designed for SAR analysis was the network-like similarity graph (NSG) 3, a precursor of more generally defined chemical space networks (CSNs) 4, which are characterized using statistical concepts from the interdisciplinary field of network science 5. As SAR-oriented network representations, NSGs provide immediate visual access to local communities (subsets) of active compounds with interesting SAR characteristics. A major distinguishing feature of different CSNs is the way in which molecular similarity relationships are established 5. The use of alternative similarity measures often changes local and global network properties of CSNs 5. When numerical similarity measures are used, pairwise compound comparisons yield a similarity matrix that contains similarity values for all compound pairs in a data set. The application of a similarity threshold value then transforms the similarity matrix into an adjacency matrix, which serves as input for layout algorithms to generate a graphical representation 6. In fact, network appearance is often strongly influenced by chosen layout algorithms. Conventional chemical space representations used in chemoinformatics are mostly generated on the basis of vectors of numerical descriptors. The resulting coordinate-based space representations are multi- or high-dimensional, with each chosen descriptor adding another dimension to the space. In such coordinate-based spaces, compound positions are unambiguously defined and so are distances between compounds that are quantified as a measure of dissimilarity, i.e. the larger the distance is, the more dissimilar the compounds are. By contrast, CSNs have become a paradigm of coordinate-free chemical space representations, which are entirely determined by pairwise similarity relationships 4,5. If substructure-based similarity measures are employed, binary relationships are obtained (i.e. two compounds are either similar or not); if similarity threshold values are applied to numerical measures, pairs of compounds reaching the threshold are classified as similar (and appear in the adjacency matrix). Hence, distance relationships between compounds are typically not considered in coordinate-free chemical space representations. In this work, we introduce a novel layout for CSNs that does not depend on chosen threshold values, but takes distances derived from pairwise similarity values into account. Thus, in contrast to currently available CSNs, distances between compounds and communities in the resulting networks become chemically relevant (at least with respect to chosen descriptors), which further increases the information content of these representations. Methods Data sets For network design, one large and three small compound sets (active against human targets with defined equilibrium constants) were taken from ChEMBL (version 21) (https://www.ebi.ac.uk/ chembl/) 7, as reported in Table 1. We note that there was no specific reason to focus on these sets; many others could have been selected instead. Molecular representation and similarity metric Compounds were represented using the MACCS fingerprint 8 (consisting of 166 structural keys or patterns), which were generated using an in-house Python implementation. Pairwise similarity values were calculated using the Tanimoto coefficient (Tc) 9. Fingerprint descriptors of different design might have been selected instead, but for our proof-of-principle investigation, the relatively simple MACCS fingerprint was readily sufficient. Similarity vs. distance Pairwise similarity values were transformed into distances using the formula distance = 1 CDF(similarity) where CDF is the cumulative distribution function for an assumed normal distribution. For each compound set, the mean and standard deviation were calculated from its pairwise similarity values. The CDF was used to emphasize compound pairs with large Tc values and de-emphasize pairs with small values compared to a linear relationship. Network layouts Alternative CSN layouts were generated with in-house Java programs based upon the JUNG library (http://jung.sourceforge. net/doc/jung_journal.pdf). Please also see the Data availability section. Table 1. Compound sets. ID Target set # CPDs 11638 MAP kinase ERK2 inhibitors 90 222 Glutamate [NMDA] receptor subunit ε 2 ligands 100476 Apoptosis regulator Bcl-W inhibitors 48 51 Serotonin 1a (5-HT1a) receptor ligands 59 1680 ID is the ChEMBL target identifier and # CPDs means number of compounds. Page 2 of 10

Fruchterman-Reingold. The Fruchterman-Reingold (FR) algorithm 10 has so far consistently been used for NSGs 3 and CSNs 5. FR is a force-directed algorithm that brings together subsets of densely connected objects and separates different subsets from each other through repulsion (until equilibrium positions are obtained). Only similarity values reaching a pre-defined threshold are considered in FR layout construction (all other similarity values are ignored). In FR-based network views, distances between compounds have no chemical meaning. Kamada-Kawai. The Kamada-Kawai (KK) algorithm 11, adapted herein for CSN design, is also a force-directed layout method. However, KK uses all distances derived from similarity values as input, and optimizes (threshold-independent) edge lengths with respect to inter-compound distances. Thus, the KK approach incorporates distance relationships into network layouts. In principle, KK-based networks are completely connected. Thus, edges between distant compounds might be omitted for clarity. Although all similarity values and corresponding distance relationships are considered for network construction, for selective edge display, similarity threshold values can also be applied. As similarity-based compound networks, KK network representations are covered by the general definition of CSNs 4,5 and are in the following also referred to as KK CSNs. Results and discussion Kamada-Kawai network design The characteristic feature of the KK approach is that it takes distances derived from all pairwise similarity values quantitatively into account during network construction. The resulting layout reflects relative compound distances, which principally increases the chemical information contained in KK CSNs compared to thresholddependent FR CSNs. Independent of the KK network structure, which remains constant, edges in KK CSNs can be selectively displayed at varying similarity threshold values to optimize the clarity of the presentation. similar data points with varying inter-subset similarity values (Table 2), whereas subset D consisted of dissimilar data points (singletons). The KK CSN of this model data set is shown in Figure 1. All three subsets of similar data points formed separate clusters in the network, whereas data points from subset D were widely distributed. Furthermore, clusters of subsets A and B, which displayed largest inter-subset similarity values (Table 2), were located close to each other and removed from the less similar subset C. Moreover, the KK CSN also correctly accounted for the smaller distance between A and C compared to B and C. Thus, the KK CSN incorporated for various distance relationships present in the model set; an encouraging finding. Kamada-Kawai networks for different sets of bioactive compounds Figure 2 shows KK CSNs for inhibitors of MAP kinase ERK2 and glutamate [NMDA] receptor subunit ε2 (data sets 11638 and 222, respectively) (Table 1). In each case, edges were selectively displayed at three different similarity threshold values, which enabled viewing edge distributions on a sliding scale. The KK CSN of the MAP kinase inhibitor set 11638 revealed a clear clustering of similar compounds with comparably high or low potency, corresponding to the presence of locally continuous SARs 1. By contrast, the KK CSN of the set of ligands of glutamate [NMDA] receptor subunit ε2 revealed a cluster of highly similar compounds with large potency variations, corresponding to a high degree of local SAR discontinuity 1. This cluster was distant from other compounds of this set, consistent with the presence of unique structural features. Kamada-Kawai network of a model data set For an initial proof-of-principle assessment, a model data set was generated by combining four subsets (A D) of five hypothetical data points, each with well-defined intra-set similarity value ranges, as reported in Table 2. Subsets A C contained highly Table 2. Similarity relationships in a model data set. A B C D A 1.0-0.9 0.8-0.7 0.6-0.5 0.1-0.0 B 0.8-0.7 1.0-0.9 0.4-0.3 0.1-0.0 C 0.6-0.5 0.4-0.3 1.0-0.9 0.1-0.0 D 0.1-0.0 0.1-0.0 0.1-0.0 0.1-0.0 For each subset of compounds in the model data set, intra-set (diagonal) and inter-set MACCS Tc value ranges are given. Figure 1. Kamada-Kawai network of a model data set. Shown is the KK CSN of the model data set according to Table 2. Data points are colored on the basis of compound subset membership: A, blue; B, red; C, green; D, gray. Page 3 of 10

Figure 2. Chemical space networks with compound distance relationships. KK CSNs are displayed for two data sets (11638 and 222 according to Table 1) at three similarity threshold values of 0.8, 0.7, and 0.6, respectively. Nodes are colored on the basis of potency values applying a color gradient from green (highest potency) over yellow (intermediate) to red (lowest potency). Page 4 of 10

Because edge densities can be selectively displayed, KK CSNs generated for larger compound data sets can be readily analyzed. In addition, for SAR analysis, one may initially select a high threshold value to primarily focus on intra-cluster relationships and then gradually decrease the threshold to concentrate more on inter-cluster connections, as illustrated in Figure 2. Comparison of Kawada-Kawai and Fruchterman-Reingold networks Figure 3 compares the KK and FR CSNs for a set of inhibitors of the apoptosis regulator Bcl-W (data set 100476), revealing the presence of distinct layouts. In the KK CSN a larger cluster of similar and mostly weakly potent compounds emerged that was distant from other data set compounds. The corresponding FR CSN provided a completely different view of the compound set with several clusters that were essentially evenly distributed across the layout (consistent with its threshold-dependent force-directed design). For each of these clusters, a corresponding cluster was also identified in the KK CSN. In three cases, the corresponding compounds were so similar and the resulting distances so small that these clusters needed to be magnified for a detailed inspection, as shown in Figure 4a. Hence, the KK and FR CSNs also provided complementary network views of the data set. The scatter plots in Figure 3 reveal that there was no correlation between similarity values and network distances in the FR CSN, consistent with its design principles. By contrast, with a correlation coefficient of -0.79, significant inverse correlation (i.e. large similarity values corresponding to small distances) was observed for the KK CSN, which was largely determined by compound pairs with similarity values greater than 0.5. For small similarity values, correlation was only weak. This observation was consistent with the use of the CDF in the distance function, which emphasized distance relationships between similar compounds, as discussed above. For data sets 222 and 11638 (Figure 2), KK CSNs yielded correlation coefficients of -0.84 and -0.88, respectively. Figure 3. Comparison of Kamada-Kawai and Fruchterman-Reingold layouts. For data set 100476, KK and FR CSNs are compared at a similarity threshold value of 0.8 for selective edge display (KK) and network generation (FR). Nodes are colored according to Figure 2. At the bottom, similarity values and corresponding network distances of all compound pairs are compared in scatter plots and correlation coefficients are reported. Page 5 of 10

Figure 4. Comparison of compound communities. In (a), corresponding compound communities are highlighted in the KK and FR CSNs from Figure 3 and enlarged. Compounds in each community are numbered. In (b), compounds forming the top cluster in (a) are shown. Page 6 of 10

Comparison of compound communities and series In Figure 4a, corresponding compound communities in KK and FR CSNs are compared in detail. FR CSN clusters contain edges of comparable length and have similar topology, which is a characteristic feature of this layout. By contrast, KK CSN clusters display different topologies and contain edges of different length that further differentiate intra-cluster similarity relationships and position similar compounds closely together. For example, compounds 3, 4, and 5 from the cluster at the top in Figure 4a only differ by the (ortho, meta, or para) position of a benzene ring and are more similar to each other than to compounds 1, 2, 6, and 7 that have different substituents (Figure 4b). Figure 5 shows a KK CSN representation for three analog series (A, B, and C) that were extracted from compounds active against the serotonin 1a receptor (data set 51). Series A and B had chemically related core structures, whereas the core of series C was distinct from A and B. In the KK CSN, the three series formed communities that were separated from each other. Consistent with the structural relationship between their cores, series A and B were positioned closer to each other than to series C. A single compound from series A was found to form a bridge between the communities of A and B. This compound contained a cyclobutyl substituent at R1 and thus closely resembled the core of series B. Taken together, these observations indicated that the KK CSN captured Figure 5. Exemplary compound series. Shown are three analog series from the KK CSN of data set 51. For clarity, a similarity threshold value of 0.88 was applied for edge display. Each analog series is encircled and its common core structure is displayed. Compounds in each series were distinguished by substituents at a single site (R1). Page 7 of 10

similarity relationships between these analog series in a meaningful way. Conclusions We have introduced an approach to incorporate compound distance relationships into CSNs that are coordinate-free representations of chemical space. For this purpose, the KK algorithm was adapted, which takes into account all inter-compound distances during network construction and does not depend on chosen similarity threshold values, in contrast to the FR algorithm. As such, KK networks also represent the first threshold-independent CSNs for numerical similarity measures, which further extends the current CSN spectrum. Initial results obtained for KK CSNs were encouraging, as demonstrated by the study of a model data set, for which subset relationships were correctly reproduced. Informative KK CSNs were also obtained for sets of bioactive compounds. Furthermore, we have shown that KK and FR CSNs may provide complementary representations that make it possible to view and compare compound communities in different ways. KK CSNs were also found to capture chemical relationships between analog series, which provided an advantage compared to FR CSNs. In summary, the results of our proof-of-principle investigation suggest that KK CSNs should be of considerable interest for further exploring biologically relevant chemical space. Data availability The data sets used in this study are freely available in ChEMBL (https://www.ebi.ac.uk/chembl/) via the identifiers reported Table 1. The NSG (FR CSN) software is freely available as a part of the SARANEA program suite 12 in an open access deposition (DOI: 10.12688/f1000research.3713.1) 13. The implementation can be adapted to generate KK CSNs. Author contributions AVL and JB conceived the study, AVL carried out the analysis, AVL and JB wrote the manuscript. Competing interests No competing interests were disclosed. Grant information The author(s) declared that no grants were involved in supporting this work. Acknowledgements We thank Dagmar Stumpfe for help with illustrations. References 1. Peltason L, Bajorath J: Systematic computational analysis of structure-activity relationships: concepts, challenges and recent advances. Future Med Chem. 2009; 1(3): 451 466. 2. Stumpfe D, Bajorath J: Methods for SAR visualization. RSC Adv. 2012; 2(2): 369 378. Publisher Full Text 3. Wawer M, Peltason L, Weskamp N, et al.: Structure-activity relationship anatomy by network-like similarity graphs and local structure-activity relationship indices. J Med Chem. 2008; 51(19): 6075 6084. 4. Maggiora GM, Bajorath J: Chemical space networks: a powerful new paradigm for the description of chemical space. J Comput Aided Mol Des. 2014; 28(8): 795 802. 5. Vogt M, Stumpfe D,Maggiora GM, et al.: Lessons learned from the design of chemical space networks and opportunities for new applications. J Comput Aided Mol Des. 2016; 30(3): 191 208. 6. Brandes U: Drawing on physical analogies. In: Drawing graphs: methods and models. Kaufmann M, Wagner D (eds.); Springer Berlin Heidelberg. 2001; 71 86. Publisher Full Text 7. Gaulton A, Bellis LJ, Bento AP, et al.: ChEMBL: a large-scale bioactivity database for drug discovery. Nucleic Acids Res. 2012; 40(Database issue): D1100 D1107. Free Full Text 8. Durant JL, Leland BA, Henry DR, et al.: Reoptimization of MDL keys for use in drug discovery. J Chem Inf Comput Sci. 2002; 42(6): 1273 1280. 9. Willet P, Barnard J, Downs GM: Chemical similarity searching. J Chem Inf Comp Sci. 1998; 38(6): 983 996. Publisher Full Text 10. Fruchterman TM, Reingold EM: Graph drawing by force-directed placement. Softw Pract Exp. 1991; 21(11): 1129 1164. Publisher Full Text 11. Kamada T, Kawai S: An algorithm for drawing general undirected graphs. Inform Process Lett. 1989; 31(1): 7 15. Publisher Full Text 12. Lounkine E, Wawer M, Wassermann AM, et al.: SARANEA: a freely available program to mine structure-activity and structure-selectivity relationship information in compound data sets. J Chem Inf Model. 2010; 50(1): 68 78. 13. Hu Y, Bajorath J: Follow up: Compound data sets and software tools for chemoinformatics and medicinal chemistry applications: update and data transfer [version 1; referees: 3 approved]. F1000Res. 2014; 3: 69. Free Full Text Page 8 of 10

Open Peer Review Current Referee Status: Version 2 Referee Report 30 November 2016 doi:10.5256/f1000research.11126.r18086 Alexandre Varnek Laboratory of Chemoinformatics, UMR 7140, CNRS (French National Center for Scientific Research), University of Strasbourg (UDS), Strasbourg, France Competing Interests: No competing interests were disclosed. I have read this submission. I believe that I have an appropriate level of expertise to confirm that it is of an acceptable scientific standard. Version 1 Referee Report 22 November 2016 doi:10.5256/f1000research.10797.r17426 Gerhard Hessler Sanofi-Aventis Deutschland GmbH., Frankfurt, Germany Chemical space networks are an interesting method to visualize chemical space and to analyse SAR of chemical series. Recently, behavior of CSN dependent on data sets and parameter settings have been carefully analysed esp. by Prof. Bajorath and co-workers. Typically the layout of CSNs is optimized for visualization purposes and thus, distances between nodes do not have a chemical meaning. Here, a method is presented, which takes into account chemical distance information in generating the network layout, which is particularly helpful in SAR analysis. The effect of the algorithm is nicely illustrated, first with a model data set and then with real SAR data sets. In addition, a comparison between distances in the graph and similarity values shows, that the design goal to reflected compound similarity in the graph distance is achieved. The publication is well written, clearly structured and adds an interesting, valuable feature to CSNs. Some small additions might be considered by the authors. Instead of referencing the target Chembl ID in the text naming the target might make reading easier. Is it possible to discuss the effect of network density on layout and SAR interpretation, esp. for larger data sets? Competing Interests: No competing interests were disclosed. Page 9 of 10

I have read this submission. I believe that I have an appropriate level of expertise to confirm that it is of an acceptable scientific standard. Referee Report 21 November 2016 doi:10.5256/f1000research.10797.r17816 Alexandre Varnek Laboratory of Chemoinformatics, UMR 7140, CNRS (French National Center for Scientific Research), University of Strasbourg (UDS), Strasbourg, France Chemical space networks (CSN) technique is an efficient way to visualize and analyze the content of chemical databases. Typically, CSNs are built using the Fruchterman-Reingold algorithm in which the distances between objects are determined for layout purposes. In this paper, for CNS construction the authors suggest to use the Kamada-Kawai algorithm providing with a graph in which the edges lengths correspond to similarity measures. Thus, CSNs obtained with the above algorithms provide with two complementary views of a chemical space. I believe that reported results are of the great interest for chemoinformatics community. The title is appropriate for the content of the article; the abstract represents a suitable summary of the work. I recommend indexing this paper as is. Competing Interests: No competing interests were disclosed. I have read this submission. I believe that I have an appropriate level of expertise to confirm that it is of an acceptable scientific standard. The benefits of publishing with F1000Research: Your article is published within days, with no editorial bias You can publish traditional articles, null/negative results, case reports, data notes and more The peer review process is transparent and collaborative Your article is indexed in PubMed after passing peer review Dedicated customer support at every stage For pre-submission enquiries, contact research@f1000.com Page 10 of 10