1
|
Choudhary R, Mahadevan R. FOCUS on NOD2: Advancing IBD Drug Discovery with a User-Informed Machine Learning Framework. ACS Med Chem Lett 2024; 15:1057-1070. [PMID: 39015268 PMCID: PMC11247655 DOI: 10.1021/acsmedchemlett.4c00148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 07/18/2024] Open
Abstract
In this study, we introduce the Framework for Optimized Customizable User-Informed Synthesis (FOCUS), a generative machine learning model tailored for drug discovery. FOCUS integrates domain expertise and uses Proximal Policy Optimization (PPO) to guide Monte Carlo Tree Search (MCTS) to efficiently explore chemical space. It generates SMILES representations of potential drug candidates, optimizing for druggability and binding efficacy to NOD2, PEP, and MCT1 receptors. The model is highly interpretive, allowing for user-feedback and expert-driven adjustments based on detailed cycle reports. Employing tools like SHAP and LIME, FOCUS provides a transparent analysis of decision-making processes, emphasizing features such as docking scores and interaction fingerprints. Comparative studies with Muramyl Dipeptide (MDP) demonstrate improved interaction profiles. FOCUS merges advanced machine learning with expert insight, accelerating the drug discovery pipeline.
Collapse
Affiliation(s)
- Ruhi Choudhary
- Department of Chemical Engineering
and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Radhakrishnan Mahadevan
- Department of Chemical Engineering
and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| |
Collapse
|
2
|
Wang L, Zhou Z, Yang X, Shi S, Zeng X, Cao D. The present state and challenges of active learning in drug discovery. Drug Discov Today 2024; 29:103985. [PMID: 38642700 DOI: 10.1016/j.drudis.2024.103985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
Active learning (AL) is an iterative feedback process that efficiently identifies valuable data within vast chemical space, even with limited labeled data. This characteristic renders it a valuable approach to tackle the ongoing challenges faced in drug discovery, such as the ever-expanding explore space and the limitations of labeled data. Consequently, AL is increasingly gaining prominence in the field of drug development. In this paper, we comprehensively review the application of AL at all stages of drug discovery, including compounds-target interaction prediction, virtual screening, molecular generation and optimization, as well as molecular properties prediction. Additionally, we discuss the challenges and prospects associated with the current applications of AL in drug discovery.
Collapse
Affiliation(s)
- Lei Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Zhenran Zhou
- Department of Computer Science, Hunan University, Changsha 410082, Hunan, China
| | - Xixi Yang
- Department of Computer Science, Hunan University, Changsha 410082, Hunan, China
| | - Shaohua Shi
- Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiangxiang Zeng
- Department of Computer Science, Hunan University, Changsha 410082, Hunan, China.
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
3
|
Zhu T, Zhang Y, Li Y, Tao T, Tao C. Contribution of molecular structures and quantum chemistry technique to root concentration factor: An innovative application of interpretable machine learning. JOURNAL OF HAZARDOUS MATERIALS 2023; 459:132320. [PMID: 37604035 DOI: 10.1016/j.jhazmat.2023.132320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/03/2023] [Accepted: 08/15/2023] [Indexed: 08/23/2023]
Abstract
Root concentration factor (RCF) is a significant parameter to characterize uptake and accumulation of hazardous organic contaminants (HOCs) by plant roots. However, complex interactions among chemicals, plant roots and soil make it challenging to identify underlying mechanisms of uptake and accumulation of HOCs. Here, nine machine learning techniques were applied to investigate major factors controlling RCF based on variable combinations of molecular descriptors (MD), MACCS fingerprints, quantum chemistry descriptors (QCD) and three physicochemical properties related to chemical-soil-plant system. Compared to models with variables including MACCS fingerprints or solitary physicochemical properties, the XGBoost-6 model developed by the variable combination of MD, QCD and three physicochemical properties achieved the most remarkable performance, with R2 of 0.977. Model interpretation achieved by permutation variable importance and partial dependence plots revealed the vital importance of HOCs lipophilicity, lipid content of plant roots, soil organic matter content, the overall deformability and the molecular dispersive ability of HOCs for regulating RCF. The integration of MD and QCD with physicochemical properties could improve our knowledge of underlying mechanisms regarding HOCs accumulation in plant roots from innovative structural perspectives. Multiple variables combination-oriented performance improvement of model can be extended to other parameters prediction in environmental risk assessment field.
Collapse
Affiliation(s)
- Tengyi Zhu
- School of Environmental Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China.
| | - Yu Zhang
- School of Environmental Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Yi Li
- School of Environmental Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Tianyun Tao
- College of Agriculture, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Cuicui Tao
- School of Environmental Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| |
Collapse
|
4
|
Park H, Hong S, Lee M, Kang S, Brahma R, Cho KH, Shin JM. AiKPro: deep learning model for kinome-wide bioactivity profiling using structure-based sequence alignments and molecular 3D conformer ensemble descriptors. Sci Rep 2023; 13:10268. [PMID: 37355672 PMCID: PMC10290719 DOI: 10.1038/s41598-023-37456-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/22/2023] [Indexed: 06/26/2023] Open
Abstract
The discovery of selective and potent kinase inhibitors is crucial for the treatment of various diseases, but the process is challenging due to the high structural similarity among kinases. Efficient kinome-wide bioactivity profiling is essential for understanding kinase function and identifying selective inhibitors. In this study, we propose AiKPro, a deep learning model that combines structure-validated multiple sequence alignments and molecular 3D conformer ensemble descriptors to predict kinase-ligand binding affinities. Our deep learning model uses an attention-based mechanism to capture complex patterns in the interactions between the kinase and the ligand. To assess the performance of AiKPro, we evaluated the impact of descriptors, the predictability for untrained kinases and compounds, and kinase activity profiling based on odd ratios. Our model, AiKPro, shows good Pearson's correlation coefficients of 0.88 and 0.87 for the test set and for the untrained sets of compounds, respectively, which also shows the robustness of the model. AiKPro shows good kinase-activity profiles across the kinome, potentially facilitating the discovery of novel interactions and selective inhibitors. Our approach holds potential implications for the discovery of novel, selective kinase inhibitors and guiding rational drug design.
Collapse
Affiliation(s)
- Hyejin Park
- AZothBio Inc., Rm. DA724 Hyundai Knowledge Industry Center, Hanam-si, Gyeonggi-do, Republic of Korea
| | - Sujeong Hong
- AZothBio Inc., Rm. DA724 Hyundai Knowledge Industry Center, Hanam-si, Gyeonggi-do, Republic of Korea
| | - Myeonghun Lee
- AZothBio Inc., Rm. DA724 Hyundai Knowledge Industry Center, Hanam-si, Gyeonggi-do, Republic of Korea
| | - Sungil Kang
- AZothBio Inc., Rm. DA724 Hyundai Knowledge Industry Center, Hanam-si, Gyeonggi-do, Republic of Korea
| | - Rahul Brahma
- School of Systems Biomedical Science, Soongsil University, Seoul, Republic of Korea
| | - Kwang-Hwi Cho
- School of Systems Biomedical Science, Soongsil University, Seoul, Republic of Korea
| | - Jae-Min Shin
- AZothBio Inc., Rm. DA724 Hyundai Knowledge Industry Center, Hanam-si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
5
|
Fassio AV, Shub L, Ponzoni L, McKinley J, O’Meara MJ, Ferreira RS, Keiser MJ, de Melo Minardi RC. Prioritizing Virtual Screening with Interpretable Interaction Fingerprints. J Chem Inf Model 2022; 62:4300-4318. [DOI: 10.1021/acs.jcim.2c00695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Alexandre V. Fassio
- São Carlos Institute of Physics, University of São Paulo, São Carlos, São Paulo 13563-120, Brazil
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Laura Shub
- Department of Pharmaceutical Chemistry, Department of Bioengineering & Therapeutic Sciences, Institute for Neurodegenerative Diseases, Kavli Institute for Fundamental Neuroscience, Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, California 94143, United States
| | - Luca Ponzoni
- Department of Pharmaceutical Chemistry, Department of Bioengineering & Therapeutic Sciences, Institute for Neurodegenerative Diseases, Kavli Institute for Fundamental Neuroscience, Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, California 94143, United States
| | - Jessica McKinley
- Gilead Sciences, Inc., Foster City, California 94404, United States
| | - Matthew J. O’Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Rafaela S. Ferreira
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Michael J. Keiser
- Department of Pharmaceutical Chemistry, Department of Bioengineering & Therapeutic Sciences, Institute for Neurodegenerative Diseases, Kavli Institute for Fundamental Neuroscience, Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, California 94143, United States
| | - Raquel C. de Melo Minardi
- Department of Computer Science, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| |
Collapse
|
6
|
Goebel GL, Qiu X, Wu P. Kinase-targeting small-molecule inhibitors and emerging bifunctional molecules. Trends Pharmacol Sci 2022; 43:866-881. [PMID: 35589447 DOI: 10.1016/j.tips.2022.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/31/2022] [Accepted: 04/11/2022] [Indexed: 02/07/2023]
Abstract
Kinases are among the most successful drug targets. To date, 72 small-molecule kinase inhibitors (SMKIs) have been approved by the US FDA, together with ~500 SMKIs in clinical trials. Although the topic has been heavily reviewed in recent years, an overview that focused on the currently approved SMKIs in combination with the emerging kinase-targeting bifunctional molecules is absent. Herein, we first provide an updated overview of the approved SMKIs, with an emphasis on their binding modes, classified in groups of type I and II ATP-competitive inhibitors, type III and IV allosteric inhibitors, and covalent inhibitors. We then highlight the novel chemical modalities in kinase targeting by using different types of proximity-inducing bifunctional molecules for kinase degradation and modifications.
Collapse
Affiliation(s)
- Georg L Goebel
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund 44227, Germany; Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund 44227, Germany; Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund 44227, Germany
| | - Xiaqiu Qiu
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund 44227, Germany; Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund 44227, Germany; Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund 44227, Germany
| | - Peng Wu
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund 44227, Germany; Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund 44227, Germany; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen DK-2100, Denmark.
| |
Collapse
|
7
|
Rodríguez-Pérez R, Miljković F, Bajorath J. Machine Learning in Chemoinformatics and Medicinal Chemistry. Annu Rev Biomed Data Sci 2022; 5:43-65. [PMID: 35440144 DOI: 10.1146/annurev-biodatasci-122120-124216] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In chemoinformatics and medicinal chemistry, machine learning has evolved into an important approach. In recent years, increasing computational resources and new deep learning algorithms have put machine learning onto a new level, addressing previously unmet challenges in pharmaceutical research. In silico approaches for compound activity predictions, de novo design, and reaction modeling have been further advanced by new algorithmic developments and the emergence of big data in the field. Herein, novel applications of machine learning and deep learning in chemoinformatics and medicinal chemistry are reviewed. Opportunities and challenges for new methods and applications are discussed, placing emphasis on proper baseline comparisons, robust validation methodologies, and new applicability domains. Expected final online publication date for the Annual Review of Biomedical Data Science, Volume 5 is August 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Raquel Rodríguez-Pérez
- Department of Life Science Informatics, B-IT (Bonn-Aachen International Center for Information Technology), Chemical Biology and Medicinal Chemistry Program Unit, LIMES (Life and Medical Sciences Institute), Rheinische Friedrich-Wilhelms-Universität, Bonn, Germany; .,Current affiliation: Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Filip Miljković
- Department of Life Science Informatics, B-IT (Bonn-Aachen International Center for Information Technology), Chemical Biology and Medicinal Chemistry Program Unit, LIMES (Life and Medical Sciences Institute), Rheinische Friedrich-Wilhelms-Universität, Bonn, Germany; .,Current affiliation: Data Science and AI, Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D AstraZeneca, Gothenburg, Sweden
| | - Jürgen Bajorath
- Department of Life Science Informatics, B-IT (Bonn-Aachen International Center for Information Technology), Chemical Biology and Medicinal Chemistry Program Unit, LIMES (Life and Medical Sciences Institute), Rheinische Friedrich-Wilhelms-Universität, Bonn, Germany;
| |
Collapse
|
8
|
Machine Learning Models for the Classification of CK2 Natural Products Inhibitors with Molecular Fingerprint Descriptors. Processes (Basel) 2021. [DOI: 10.3390/pr9112074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Casein kinase 2 (CK2) is considered an important target for anti-cancer drugs. Given the structural diversity and broad spectrum of pharmaceutical activities of natural products, numerous studies have been performed to prove them as valuable sources of drugs. However, there has been little study relevant to identifying structural factors responsible for their inhibitory activity against CK2 with machine learning methods. In this study, classification studies were conducted on 115 natural products as CK2 inhibitors. Seven machine learning methods along with six molecular fingerprints were employed to develop qualitative classification models. The performances of all models were evaluated by cross-validation and test set. By taking predictive accuracy(CA), the area under receiver operating characteristic (AUC), and (MCC)as three performance indicators, the optimal models with high reliability and predictive ability were obtained, including the Extended Fingerprint-Logistic Regression model (CA = 0.859, AUC = 0.826, MCC = 0.520) for training test andPubChem fingerprint along with the artificial neural model (CA = 0.826, AUC = 0.933, MCC = 0.628) for test set. Meanwhile, the privileged substructures responsible for their inhibitory activity against CK2 were also identified through a combination of frequency analysis and information gain. The results are expected to provide useful information for the further utilization of natural products and the discovery of novel CK2 inhibitors.
Collapse
|
9
|
Bouysset C, Fiorucci S. ProLIF: a library to encode molecular interactions as fingerprints. J Cheminform 2021; 13:72. [PMID: 34563256 PMCID: PMC8466659 DOI: 10.1186/s13321-021-00548-6] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/30/2021] [Indexed: 12/21/2022] Open
Abstract
Interaction fingerprints are vector representations that summarize the three-dimensional nature of interactions in molecular complexes, typically formed between a protein and a ligand. This kind of encoding has found many applications in drug-discovery projects, from structure-based virtual-screening to machine-learning. Here, we present ProLIF, a Python library designed to generate interaction fingerprints for molecular complexes extracted from molecular dynamics trajectories, experimental structures, and docking simulations. It can handle complexes formed of any combination of ligand, protein, DNA, or RNA molecules. The available interaction types can be fully reparametrized or extended by user-defined ones. Several tutorials that cover typical use-case scenarios are available, and the documentation is accompanied with code snippets showcasing the integration with other data-analysis libraries for a more seamless user-experience. The library can be freely installed from our GitHub repository (https://github.com/chemosim-lab/ProLIF).
Collapse
Affiliation(s)
- Cédric Bouysset
- Institut de Chimie de Nice UMR7272, Université Côte d'Azur, CNRS, Nice, France.
| | - Sébastien Fiorucci
- Institut de Chimie de Nice UMR7272, Université Côte d'Azur, CNRS, Nice, France.
| |
Collapse
|
10
|
Fischer A, Smieško M, Sellner M, Lill MA. Decision Making in Structure-Based Drug Discovery: Visual Inspection of Docking Results. J Med Chem 2021; 64:2489-2500. [PMID: 33617246 DOI: 10.1021/acs.jmedchem.0c02227] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Molecular docking is a computational method widely used in drug discovery. Due to the inherent inaccuracies of molecular docking, visual inspection of binding modes is a crucial routine in the decision making process of computational medicinal chemists. Despite its apparent importance for medicinal chemistry projects, guidelines for the visual docking pose assessment have been hardly discussed in the literature. Here, we review the medicinal chemistry literature with the aim of identifying consistent principles for visual inspection, highlighting cases of its successful application, and discussing its limitations. In this context, we conducted a survey reaching experts in both academia and the pharmaceutical industry, which also included a challenge to distinguish native from incorrect poses. We were able to collect 93 expert opinions that offer valuable insights into visually supported decision-making processes. This perspective shall motivate discussions among experienced computational medicinal chemists and guide young scientists new to the field to stratify their compounds.
Collapse
Affiliation(s)
- André Fischer
- Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland
| | - Martin Smieško
- Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland
| | - Manuel Sellner
- Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland
| | - Markus A Lill
- Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland
| |
Collapse
|
11
|
Abdelbaky I, Tayara H, Chong KT. Prediction of kinase inhibitors binding modes with machine learning and reduced descriptor sets. Sci Rep 2021; 11:706. [PMID: 33436888 PMCID: PMC7804204 DOI: 10.1038/s41598-020-80758-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 12/28/2020] [Indexed: 02/06/2023] Open
Abstract
Protein kinases are receiving wide research interest, from drug perspective, due to their important roles in human body. Available kinase-inhibitor data, including crystallized structures, revealed many details about the mechanism of inhibition and binding modes. The understanding and analysis of these binding modes are expected to support the discovery of kinase-targeting drugs. The huge amounts of data made it possible to utilize computational techniques, including machine learning, to help in the discovery of kinase-targeting drugs. Machine learning gave reasonable predictions when applied to differentiate between the binding modes of kinase inhibitors, promoting a wider application in that domain. In this study, we applied machine learning supported by feature selection techniques to classify kinase inhibitors according to their binding modes. We represented inhibitors as a large number of molecular descriptors, as features, and systematically reduced these features in a multi-step manner while trying to attain high classification accuracy. Our predictive models could satisfy both goals by achieving high accuracy while utilizing at most 5% of the modeling features. The models could differentiate between binding mode types with MCC values between 0.67 and 0.92, and balanced accuracy values between 0.78 and 0.97 for independent test sets.
Collapse
Affiliation(s)
- Ibrahim Abdelbaky
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju, 54896, South Korea.,Agricultural Research Center, Giza, 12619, Egypt.,Advanced Electronics and Information Research Center, Jeonbuk National University, Jeonju, 54896, South Korea
| | - Hilal Tayara
- School of International Engineering and Science, Jeonbuk National University, Jeonju, 54896, South Korea.
| | - Kil To Chong
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju, 54896, South Korea. .,Advanced Electronics and Information Research Center, Jeonbuk National University, Jeonju, 54896, South Korea.
| |
Collapse
|
12
|
Kanev GK, de Graaf C, Westerman BA, de Esch IJP, Kooistra AJ. KLIFS: an overhaul after the first 5 years of supporting kinase research. Nucleic Acids Res 2021; 49:D562-D569. [PMID: 33084889 PMCID: PMC7778968 DOI: 10.1093/nar/gkaa895] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 09/30/2020] [Indexed: 12/14/2022] Open
Abstract
Kinases are a prime target of drug development efforts with >60 drug approvals in the past two decades. Due to the research into this protein family, a wealth of data has been accumulated that keeps on growing. KLIFS-Kinase-Ligand Interaction Fingerprints and Structures-is a structural database focusing on how kinase inhibitors interact with their targets. The aim of KLIFS is to support (structure-based) kinase research through the systematic collection, annotation, and processing of kinase structures. Now, 5 years after releasing the initial KLIFS website, the database has undergone a complete overhaul with a new website, new logo, and new functionalities. In this article, we start by looking back at how KLIFS has been used by the research community, followed by a description of the renewed KLIFS, and conclude with showcasing the functionalities of KLIFS. Major changes include the integration of approved drugs and inhibitors in clinical trials, extension of the coverage to atypical kinases, and a RESTful API for programmatic access. KLIFS is available at the new domain https://klifs.net.
Collapse
Affiliation(s)
- Georgi K Kanev
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Department of Neurosurgery, Amsterdam University Medical Centers, Cancer Center Amsterdam, Brain Tumor Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Chris de Graaf
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abington, Cambridge, CB21 6DG, UK
| | - Bart A Westerman
- Department of Neurosurgery, Amsterdam University Medical Centers, Cancer Center Amsterdam, Brain Tumor Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Albert J Kooistra
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| |
Collapse
|
13
|
Machine learning, artificial intelligence, and data science breaking into drug design and neglected diseases. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2021. [DOI: 10.1002/wcms.1513] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
14
|
Tetko IV, Engkvist O. From Big Data to Artificial Intelligence: chemoinformatics meets new challenges. J Cheminform 2020; 12:74. [PMID: 33339533 PMCID: PMC7747384 DOI: 10.1186/s13321-020-00475-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/17/2022] Open
Abstract
The increasing volume of biomedical data in chemistry and life sciences requires development of new methods and approaches for their analysis. Artificial Intelligence and machine learning, especially neural networks, are increasingly used in the chemical industry, in particular with respect to Big Data. This editorial highlights the main results presented during the special session of the International Conference on Neural Networks organized by "Big Data in Chemistry" project and draws perspectives on the future progress of the field.
Collapse
Affiliation(s)
- Igor V Tetko
- Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Institute of Structural Biology, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
- BIGCHEM GmbH, Valerystr. 49, 85716, Unterschleißheim, Germany.
| | - Ola Engkvist
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|