51
|
Mouchlis VD, Afantitis A, Serra A, Fratello M, Papadiamantis AG, Aidinis V, Lynch I, Greco D, Melagraki G. Advances in de Novo Drug Design: From Conventional to Machine Learning Methods. Int J Mol Sci 2021; 22:1676. [PMID: 33562347 PMCID: PMC7915729 DOI: 10.3390/ijms22041676] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/31/2021] [Accepted: 01/31/2021] [Indexed: 12/11/2022] Open
Abstract
. De novo drug design is a computational approach that generates novel molecular structures from atomic building blocks with no a priori relationships. Conventional methods include structure-based and ligand-based design, which depend on the properties of the active site of a biological target or its known active binders, respectively. Artificial intelligence, including machine learning, is an emerging field that has positively impacted the drug discovery process. Deep reinforcement learning is a subdivision of machine learning that combines artificial neural networks with reinforcement-learning architectures. This method has successfully been employed to develop novel de novo drug design approaches using a variety of artificial networks including recurrent neural networks, convolutional neural networks, generative adversarial networks, and autoencoders. This review article summarizes advances in de novo drug design, from conventional growth algorithms to advanced machine-learning methodologies and highlights hot topics for further development.
Collapse
Affiliation(s)
| | - Antreas Afantitis
- Department of ChemoInformatics, NovaMechanics Ltd., Nicosia 1046, Cyprus;
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (A.S.); (M.F.); (D.G.)
- BioMEdiTech Institute, Tampere University, 33520 Tampere, Finland
| | - Michele Fratello
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (A.S.); (M.F.); (D.G.)
- BioMEdiTech Institute, Tampere University, 33520 Tampere, Finland
| | - Anastasios G. Papadiamantis
- Department of ChemoInformatics, NovaMechanics Ltd., Nicosia 1046, Cyprus;
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Vassilis Aidinis
- Institute for Bioinnovation, Biomedical Sciences Research Center Alexander Fleming, Fleming 34, 16672 Athens, Greece;
| | - Iseult Lynch
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (A.S.); (M.F.); (D.G.)
- BioMEdiTech Institute, Tampere University, 33520 Tampere, Finland
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
- Finnish Center for Alternative Methods (FICAM), Tampere University, 33520 Tampere, Finland
| | - Georgia Melagraki
- Division of Physical Sciences & Applications, Hellenic Military Academy, 16672 Vari, Greece
| |
Collapse
|
52
|
Al-Ostoot FH, Salah S, Khanum SA. Recent investigations into synthesis and pharmacological activities of phenoxy acetamide and its derivatives (chalcone, indole and quinoline) as possible therapeutic candidates. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2021. [PMCID: PMC7849228 DOI: 10.1007/s13738-021-02172-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Medicinal chemistry can rightfully be regarded as a cornerstone in the public health of our modern society that combines chemistry and pharmacology with the aim of designing and developing new pharmaceutical compounds. For this purpose, many chemical techniques as well as new computational chemistry applications are used to study the utilization of drugs and their biological effects. In the biological interface, medicinal chemistry constitutes a group of interdisciplinary sciences, as well as controlling its organic, physical and computational pillars. Therefore, medicinal chemists working to design an integrated and developing system that portends an era of novel and safe tailored drugs either by synthesizing new pharmaceuticals or to improving the processes by which existing pharmaceuticals are made. It includes researching the effects of synthetic, semi-synthetic and natural biologically active substances based on molecular interactions in terms of molecular structure with triggered functional groups or the specific physicochemical properties. The present work focuses on the literature survey of chemical diversity of phenoxy acetamide and its derivatives (Chalcone, Indole and Quinoline) in the molecular framework in order to get complete information regarding pharmacologically interesting compounds of widely different composition. From a biological and industrial point of view, this literature review may provide an opportunity for the chemists to design new derivatives of phenoxy acetamide and its derivatives that proved to be the successful agent in view of safety and efficacy to enhance life quality.
Collapse
Affiliation(s)
- Fares Hezam Al-Ostoot
- Department of Chemistry, Yuvaraja’s College, University of Mysore, Mysuru, 570 006 India
- Department of Biochemistry, Faculty of Education and Science, Al-Baydha University, Al-Baydha, Yemen
| | - Salma Salah
- Faculty of Medicine and Health Sciences, Thamar University, Dhamar, Yemen
| | - Shaukath Ara Khanum
- Department of Chemistry, Yuvaraja’s College, University of Mysore, Mysuru, 570 006 India
| |
Collapse
|
53
|
|
54
|
Bender A, Cortés-Ciriano I. Artificial intelligence in drug discovery: what is realistic, what are illusions? Part 1: Ways to make an impact, and why we are not there yet. Drug Discov Today 2020; 26:511-524. [PMID: 33346134 DOI: 10.1016/j.drudis.2020.12.009] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/07/2020] [Accepted: 12/11/2020] [Indexed: 12/30/2022]
Abstract
Although artificial intelligence (AI) has had a profound impact on areas such as image recognition, comparable advances in drug discovery are rare. This article quantifies the stages of drug discovery in which improvements in the time taken, success rate or affordability will have the most profound overall impact on bringing new drugs to market. Changes in clinical success rates will have the most profound impact on improving success in drug discovery; in other words, the quality of decisions regarding which compound to take forward (and how to conduct clinical trials) are more important than speed or cost. Although current advances in AI focus on how to make a given compound, the question of which compound to make, using clinical efficacy and safety-related end points, has received significantly less attention. As a consequence, current proxy measures and available data cannot fully utilize the potential of AI in drug discovery, in particular when it comes to drug efficacy and safety in vivo. Thus, addressing the questions of which data to generate and which end points to model will be key to improving clinically relevant decision-making in the future.
Collapse
Affiliation(s)
- Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road CB2 1EW, UK; Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
| | - Isidro Cortés-Ciriano
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD, UK.
| |
Collapse
|
55
|
Jeon W, Kim D. Autonomous molecule generation using reinforcement learning and docking to develop potential novel inhibitors. Sci Rep 2020; 10:22104. [PMID: 33328504 PMCID: PMC7744578 DOI: 10.1038/s41598-020-78537-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 11/25/2020] [Indexed: 12/16/2022] Open
Abstract
We developed a computational method named Molecule Optimization by Reinforcement Learning and Docking (MORLD) that automatically generates and optimizes lead compounds by combining reinforcement learning and docking to develop predicted novel inhibitors. This model requires only a target protein structure and directly modifies ligand structures to obtain higher predicted binding affinity for the target protein without any other training data. Using MORLD, we were able to generate potential novel inhibitors against discoidin domain receptor 1 kinase (DDR1) in less than 2 days on a moderate computer. We also demonstrated MORLD's ability to generate predicted novel agonists for the D4 dopamine receptor (D4DR) from scratch without virtual screening on an ultra large compound library. The free web server is available at http://morld.kaist.ac.kr .
Collapse
Affiliation(s)
- Woosung Jeon
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Dongsup Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
56
|
The Potential of 19F NMR Application in GPCR Biased Drug Discovery. Trends Pharmacol Sci 2020; 42:19-30. [PMID: 33250272 DOI: 10.1016/j.tips.2020.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/16/2020] [Accepted: 11/02/2020] [Indexed: 01/14/2023]
Abstract
Although structure-based virtual drug discovery is revolutionizing the conventional high-throughput cell-based screening system, its limitation is obvious, together with other critical challenges. In particular, the resolved static snapshots fail to represent a full free-energy landscape due to homogenization in structural determination processing. The loss of conformational heterogeneity and related functional diversity emphasize the necessity of developing an approach that can fill this space. In this regard, NMR holds undeniable potential. However, outstanding questions regarding the NMR application remain. This review summarizes the limitations of current drug discovery and explores the potential of 19F NMR in establishing a conformation-guided drug screening system, advancing the cell- and structure-based discovery strategy for G protein-coupled receptor (GPCR) biased drug screening.
Collapse
|
57
|
Lo SC, Xie ZR, Chang KY. Structural and Functional Enrichment Analyses for Antimicrobial Peptides. Int J Mol Sci 2020; 21:E8783. [PMID: 33233636 PMCID: PMC7699717 DOI: 10.3390/ijms21228783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/28/2022] Open
Abstract
Whether there is any inclination between structures and functions of antimicrobial peptides (AMPs) is a mystery yet to be unraveled. AMPs have various structures associated with many different antimicrobial functions, including antibacterial, anticancer, antifungal, antiparasitic and antiviral activities. However, none has yet reported any antimicrobial functional tendency within a specific category of protein/peptide structures nor any structural tendency of a specific antimicrobial function with respect to AMPs. Here, we examine the relationships between structures categorized by three structural classification methods (CATH, SCOP, and TM) and seven antimicrobial functions with respect to AMPs using an enrichment analysis. The results show that antifungal activities of AMPs were tightly related to the two-layer sandwich structure of CATH, the knottin fold of SCOP, and the first structural cluster of TM. The associations with knottin and TM Cluster 1 even sustained through the AMPs with a low sequence identity. Moreover, another significant mutual enrichment was observed between the third cluster of TM and anti-Gram-positive-bacterial/anti-Gram-negative-bacterial activities. The findings of the structure-function inclination further our understanding of AMPs and could help us design or discover new therapeutic potential AMPs.
Collapse
Affiliation(s)
- Sheng C. Lo
- Computational Biology Laboratory, Department of Computer Science and Engineering, National Taiwan Ocean University, Keelung 202, Taiwan;
| | - Zhong-Ru Xie
- School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA 30602, USA;
| | - Kuan Y. Chang
- Computational Biology Laboratory, Department of Computer Science and Engineering, National Taiwan Ocean University, Keelung 202, Taiwan;
| |
Collapse
|
58
|
Liu Z, Huang D, Zheng S, Song Y, Liu B, Sun J, Niu Z, Gu Q, Xu J, Xie L. Deep learning enables discovery of highly potent anti-osteoporosis natural products. Eur J Med Chem 2020; 210:112982. [PMID: 33158578 DOI: 10.1016/j.ejmech.2020.112982] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/16/2020] [Accepted: 09/24/2020] [Indexed: 01/13/2023]
Abstract
A pre-trained self-attentive message passing neural network (P-SAMPNN) model was developed based on our anti-osteoclastogenesis dataset for virtual screening purpose. Validation processes proved that P-SAMPNN model was significantly superior to the other base line models. A commercially available natural product library was virtually screened by the P-SAMPNN model and resulted in confirmed 5 hits from 10 selected virtual hits. Among the confirmed hits, compounds AP-123/40765213 and AE-562/43462182 are the nanomolar inhibitors against osteoclastogenesis with a new scaffold. Further studies indicate that AP-123/40765213 and AE-562/43462182 significantly suppress the mRNA expression of RANK and downregulate the expressions of osteoclasts-related genes Ctsk, Nfatc1, and Tracp. Our work demonstrated that P-SAMPNN method can guide phenotype-based drug discovery.
Collapse
Affiliation(s)
- Zhihong Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Dane Huang
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; Guangdong Province Engineering Technology Research Institute of T.C.M., Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, 510095, China
| | - Shuangjia Zheng
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; School of Data and Computer Science, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ying Song
- School of Data and Computer Science, Sun Yat-sen University, Guangzhou, 510006, China
| | - Bingdong Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Jingyuan Sun
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhangming Niu
- Aladdin Healthcare Technologies Ltd., 24-26, Baltic Street West, London EC1Y OUR, UK
| | - Qiong Gu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Jun Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; School of Biotechnology and Health Sciences, Wuyi University, 99 Yingbin Road, Jiangmen, 529020, China.
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China.
| |
Collapse
|
59
|
Leguy J, Cauchy T, Glavatskikh M, Duval B, Da Mota B. EvoMol: a flexible and interpretable evolutionary algorithm for unbiased de novo molecular generation. J Cheminform 2020; 12:55. [PMID: 33431049 PMCID: PMC7494000 DOI: 10.1186/s13321-020-00458-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/31/2020] [Indexed: 11/24/2022] Open
Abstract
The objective of this work is to design a molecular generator capable of exploring known as well as unfamiliar areas of the chemical space. Our method must be flexible to adapt to very different problems. Therefore, it has to be able to work with or without the influence of prior data and knowledge. Moreover, regardless of the success, it should be as interpretable as possible to allow for diagnosis and improvement. We propose here a new open source generation method using an evolutionary algorithm to sequentially build molecular graphs. It is independent of starting data and can generate totally unseen compounds. To be able to search a large part of the chemical space, we define an original set of 7 generic mutations close to the atomic level. Our method achieves excellent performances and even records on the QED, penalised logP, SAscore, CLscore as well as the set of goal-directed functions defined in GuacaMol. To demonstrate its flexibility, we tackle a very different objective issued from the organic molecular materials domain. We show that EvoMol can generate sets of optimised molecules having high energy HOMO or low energy LUMO, starting only from methane. We can also set constraints on a synthesizability score and structural features. Finally, the interpretability of EvoMol allows for the visualisation of its exploration process as a chemically relevant tree. ![]()
Collapse
Affiliation(s)
- Jules Leguy
- Laboratoire LERIA, UNIV Angers, SFR MathSTIC, 2 Bd Lavoisier, 49045, Angers, France
| | - Thomas Cauchy
- Laboratoire MOLTECH-Anjou, UMR CNRS 6200, UNIV Angers, SFR MATRIX, 2 Bd Lavoisier, 49045, Angers, France.
| | - Marta Glavatskikh
- Laboratoire LERIA, UNIV Angers, SFR MathSTIC, 2 Bd Lavoisier, 49045, Angers, France.,Laboratoire MOLTECH-Anjou, UMR CNRS 6200, UNIV Angers, SFR MATRIX, 2 Bd Lavoisier, 49045, Angers, France
| | - Béatrice Duval
- Laboratoire LERIA, UNIV Angers, SFR MathSTIC, 2 Bd Lavoisier, 49045, Angers, France
| | - Benoit Da Mota
- Laboratoire LERIA, UNIV Angers, SFR MathSTIC, 2 Bd Lavoisier, 49045, Angers, France.
| |
Collapse
|
60
|
Domenico A, Nicola G, Daniela T, Fulvio C, Nicola A, Orazio N. De Novo Drug Design of Targeted Chemical Libraries Based on Artificial Intelligence and Pair-Based Multiobjective Optimization. J Chem Inf Model 2020; 60:4582-4593. [PMID: 32845150 DOI: 10.1021/acs.jcim.0c00517] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Artificial intelligence and multiobjective optimization represent promising solutions to bridge chemical and biological landscapes by addressing the automated de novo design of compounds as a result of a humanlike creative process. In the present study, we conceived a novel pair-based multiobjective approach implemented in an adapted SMILES generative algorithm based on recurrent neural networks for the automated de novo design of new molecules whose overall features are optimized by finding the best trade-offs among relevant physicochemical properties (MW, logP, HBA, HBD) and additional similarity-based constraints biasing specific biological targets. In this respect, we carried out the de novo design of chemical libraries targeting neuraminidase, acetylcholinesterase, and the main protease of severe acute respiratory syndrome coronavirus 2. Several quality metrics were employed to assess drug-likeness, chemical feasibility, diversity content, and validity. Molecular docking was finally carried out to better evaluate the scoring and posing of the de novo generated molecules with respect to X-ray cognate ligands of the corresponding molecular counterparts. Our results indicate that artificial intelligence and multiobjective optimization allow us to capture the latent links joining chemical and biological aspects, thus providing easy-to-use options for customizable design strategies, which are especially effective for both lead generation and lead optimization. The algorithm is freely downloadable at https://github.com/alberdom88/moo-denovo and all of the data are available as Supporting Information.
Collapse
Affiliation(s)
- Alberga Domenico
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Via E. Orabona, 4, I-70126 Bari, Italy
| | - Gambacorta Nicola
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Via E. Orabona, 4, I-70126 Bari, Italy
| | - Trisciuzzi Daniela
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Via E. Orabona, 4, I-70126 Bari, Italy.,Molecular Horizon srl, Via Montelino 32, 06084 Bettona, Italy
| | - Ciriaco Fulvio
- Dipartimento di Chimica, Università degli Studi di Bari "Aldo Moro", Via E. Orabona, 4, I-70126 Bari, Italy
| | - Amoroso Nicola
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Via E. Orabona, 4, I-70126 Bari, Italy
| | - Nicolotti Orazio
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Via E. Orabona, 4, I-70126 Bari, Italy
| |
Collapse
|
61
|
Arya H, Coumar MS. Design of novel ROCK inhibitors using fragment-based de novo drug design approach. J Mol Model 2020; 26:249. [PMID: 32829478 DOI: 10.1007/s00894-020-04493-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/30/2020] [Indexed: 12/01/2022]
Abstract
Rho-associated coiled-coil protein kinase (ROCK) is playing a vital role in the regulation of key cellular events and also responsible for causing several pathological conditions such as cancer, hypertension, Alzheimer's, cerebral vasospasm, and cardiac stroke. Therefore, it has attracted us to target ROCK protein as a potential therapeutic target for combating various diseases. Consequently, we investigated the active site of ROCK I protein and designed novel leads against the target using the de novo evolution drug design approach. Caffeic acid (an aglycone of acteoside) as a scaffold and fragments from 336 reported ROCK inhibitors were used for the design of novel leads. Multiple copy simultaneous search docking was used to identify the suitable fragments to be linked with the scaffold. Basic medicinal chemistry rules, coupled with structural insights generated by docking, led to the design of 7a, 8a, 9a, and 10a as potential ROCK I inhibitors. The designed leads showed better binding than the approved drug fasudil and also interacted with the key hinge region residue Met156 of ROCK I. Further, molecular dynamics (MD) simulation revealed that the protein-ligand complexes were stable and maintained the hydrogen bond with Met156 throughout the MD run. The promising in silico outcomes suggest that the designed compounds could be suitable anti-cancer leads that need to be synthesized and tested in various cancer cell lines. Graphical abstract.
Collapse
Affiliation(s)
- Hemant Arya
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry, 605014, India
| | - Mohane Selvaraj Coumar
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry, 605014, India.
| |
Collapse
|
62
|
Vanhaelen Q, Lin YC, Zhavoronkov A. The Advent of Generative Chemistry. ACS Med Chem Lett 2020; 11:1496-1505. [PMID: 32832015 PMCID: PMC7429972 DOI: 10.1021/acsmedchemlett.0c00088] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Generative adversarial networks (GANs), first published in 2014, are among the most important concepts in modern artificial intelligence (AI). Bridging deep learning and game theory, GANs are used to generate or "imagine" new objects with desired properties. Since 2016, multiple GANs with reinforcement learning (RL) have been successfully applied in pharmacology for de novo molecular design. Those techniques aim at a more efficient use of the data and a better exploration of the chemical space. We review recent advances for the generation of novel molecules with desired properties with a focus on the applications of GANs, RL, and related techniques. We also discuss the current limitations and challenges in the new growing field of generative chemistry.
Collapse
Affiliation(s)
- Quentin Vanhaelen
- Insilico
Medicine Hong Kong Ltd, Pak Shek Kok, New Territories, Hong Kong
| | - Yen-Chu Lin
- Insilico
Medicine Hong Kong Ltd, Pak Shek Kok, New Territories, Hong Kong
- Insilico
Taiwan, Taipei City 115, Taiwan, R.O.C
| | - Alex Zhavoronkov
- Insilico
Medicine Hong Kong Ltd, Pak Shek Kok, New Territories, Hong Kong
| |
Collapse
|
63
|
Schneider P, Welin M, Svensson B, Walse B, Schneider G. Virtual Screening and Design with Machine Intelligence Applied to Pim-1 Kinase Inhibitors. Mol Inform 2020; 39:e2000109. [PMID: 33448694 PMCID: PMC7539333 DOI: 10.1002/minf.202000109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/17/2020] [Indexed: 12/17/2022]
Abstract
Ligand-based virtual screening of large compound collections, combined with fast bioactivity determination, facilitate the discovery of bioactive molecules with desired properties. Here, chemical similarity based machine learning and label-free differential scanning fluorimetry were used to rapidly identify new ligands of the anticancer target Pim-1 kinase. The three-dimensional crystal structure complex of human Pim-1 with ligand bound revealed an ATP-competitive binding mode. Generative de novo design with a recurrent neural network additionally suggested innovative molecular scaffolds. Results corroborate the validity of the chemical similarity principle for rapid ligand prototyping, suggesting the complementarity of similarity-based and generative computational approaches.
Collapse
Affiliation(s)
- Petra Schneider
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland.,inSili.com GmbH, Segantinisteig 3, 8049, Zurich, Switzerland
| | - Martin Welin
- SARomics Biostructures AB, Medicon Village, SE-223 81, Lund, Sweden
| | - Bo Svensson
- SARomics Biostructures AB, Medicon Village, SE-223 81, Lund, Sweden
| | - Björn Walse
- SARomics Biostructures AB, Medicon Village, SE-223 81, Lund, Sweden
| | - Gisbert Schneider
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| |
Collapse
|
64
|
Shinde PB, Bhowmick S, Alfantoukh E, Patil PC, Wabaidur SM, Chikhale RV, Islam MA. De novo design based identification of potential HIV-1 integrase inhibitors: A pharmacoinformatics study. Comput Biol Chem 2020; 88:107319. [PMID: 32801062 DOI: 10.1016/j.compbiolchem.2020.107319] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/10/2020] [Accepted: 06/22/2020] [Indexed: 12/30/2022]
Abstract
In the present study, pharmacoinformatics paradigms include receptor-based de novo design, virtual screening through molecular docking and molecular dynamics (MD) simulation are implemented to identify novel and promising HIV-1 integrase inhibitors. The de novodrug/ligand/molecule design is a powerful and effective approach to design a large number of novel and structurally diverse compounds with the required pharmacological profiles. A crystal structure of HIV-1 integrase bound with standard inhibitor BI-224436 is used and a set of 80,000 compounds through the de novo approach in LigBuilder is designed. Initially, a number of criteria including molecular docking, in-silico toxicity and pharmacokinetics profile assessments are implied to reduce the chemical space. Finally, four de novo designed molecules are proposed as potential HIV-1 integrase inhibitors based on comparative analyses. Notably, strong binding interactions have been identified between a few newly identified catalytic amino acid residues and proposed HIV-1 integrase inhibitors. For evaluation of the dynamic stability of the protein-ligand complexes, a number of parameters are explored from the 100 ns MD simulation study. The MD simulation study suggested that proposed molecules efficiently retained their molecular interaction and structural integrity inside the HIV-1 integrase. The binding free energy is calculated through the Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) approach for all complexes and it also explains their thermodynamic stability. Hence, proposed molecules through de novo design might be critical to inhibiting the HIV-1 integrase.
Collapse
Affiliation(s)
- Pooja Balasaheb Shinde
- Department of Bioinformatics, Rajiv Gandhi Institute of IT and Biotechnology, Bharati Vidyapeeth Deemed University, Pune-Satara Road, Pune, India
| | - Shovonlal Bhowmick
- Department of Chemical Technology, University of Calcutta, 92, A.P.C. Road, Kolkata, 700009, India
| | - Etidal Alfantoukh
- Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Pritee Chunarkar Patil
- Department of Bioinformatics, Rajiv Gandhi Institute of IT and Biotechnology, Bharati Vidyapeeth Deemed University, Pune-Satara Road, Pune, India
| | - Saikh Mohammad Wabaidur
- Department of Chemistry P.O. Box 2455, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Rupesh V Chikhale
- School of Pharmacy, University of East Anglia, Norwich, United Kingdom
| | - Md Ataul Islam
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PL, United Kingdom; School of Health Sciences, University of Kwazulu-Natal, Westville Campus, Durban, South Africa; Department of Chemical Pathology, Faculty of Health Sciences, University of Pretoria and National Health Laboratory Service Tshwane Academic Division, Pretoria, South Africa.
| |
Collapse
|
65
|
Coley CW, Eyke NS, Jensen KF. Autonomous Discovery in the Chemical Sciences Part I: Progress. Angew Chem Int Ed Engl 2020; 59:22858-22893. [DOI: 10.1002/anie.201909987] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Connor W. Coley
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Natalie S. Eyke
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Klavs F. Jensen
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| |
Collapse
|
66
|
Coley CW, Eyke NS, Jensen KF. Autonome Entdeckung in den chemischen Wissenschaften, Teil I: Fortschritt. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201909987] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Connor W. Coley
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Natalie S. Eyke
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Klavs F. Jensen
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| |
Collapse
|
67
|
Friederich P, Dos Passos Gomes G, De Bin R, Aspuru-Guzik A, Balcells D. Machine learning dihydrogen activation in the chemical space surrounding Vaska's complex. Chem Sci 2020; 11:4584-4601. [PMID: 33224459 PMCID: PMC7659707 DOI: 10.1039/d0sc00445f] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/06/2020] [Indexed: 12/15/2022] Open
Abstract
Homogeneous catalysis using transition metal complexes is ubiquitously used for organic synthesis, as well as technologically relevant in applications such as water splitting and CO2 reduction. The key steps underlying homogeneous catalysis require a specific combination of electronic and steric effects from the ligands bound to the metal center. Finding the optimal combination of ligands is a challenging task due to the exceedingly large number of possibilities and the non-trivial ligand-ligand interactions. The classic example of Vaska's complex, trans-[Ir(PPh3)2(CO)(Cl)], illustrates this scenario. The ligands of this species activate iridium for the oxidative addition of hydrogen, yielding the dihydride cis-[Ir(H)2(PPh3)2(CO)(Cl)] complex. Despite the simplicity of this system, thousands of derivatives can be formulated for the activation of H2, with a limited number of ligands belonging to the same general categories found in the original complex. In this work, we show how DFT and machine learning (ML) methods can be combined to enable the prediction of reactivity within large chemical spaces containing thousands of complexes. In a space of 2574 species derived from Vaska's complex, data from DFT calculations are used to train and test ML models that predict the H2-activation barrier. In contrast to experiments and calculations requiring several days to be completed, the ML models were trained and used on a laptop on a time-scale of minutes. As a first approach, we combined Bayesian-optimized artificial neural networks (ANN) with features derived from autocorrelation and deltametric functions. The resulting ANNs achieved high accuracies, with mean absolute errors (MAE) between 1 and 2 kcal mol-1, depending on the size of the training set. By using a Gaussian process (GP) model trained with a set of selected features, including fingerprints, accuracy was further enhanced. Remarkably, this GP model minimized the MAE below 1 kcal mol-1, by using only 20% or less of the data available for training. The gradient boosting (GB) method was also used to assess the relevance of the features, which was used for both feature selection and model interpretation purposes. Features accounting for chemical composition, atom size and electronegativity were found to be the most determinant in the predictions. Further, the ligand fragments with the strongest influence on the H2-activation barrier were identified.
Collapse
Affiliation(s)
- Pascal Friederich
- Chemical Physics Theory Group , Department of Chemistry , University of Toronto , Toronto , Ontario M5S 3H6 , Canada
- Institute of Nanotechnology , Karlsruhe Institute of Technology , Hermann-von-Helmholtz-Platz 1 , 76344 Eggenstein-Leopoldshafen , Germany
- Department of Computer Science , University of Toronto , 214 College St. , Toronto , Ontario M5T 3A1 , Canada
| | - Gabriel Dos Passos Gomes
- Chemical Physics Theory Group , Department of Chemistry , University of Toronto , Toronto , Ontario M5S 3H6 , Canada
- Department of Computer Science , University of Toronto , 214 College St. , Toronto , Ontario M5T 3A1 , Canada
| | - Riccardo De Bin
- Department of Mathematics , University of Oslo , P. O. Box 1053, Blindern , N-0316 , Oslo , Norway
| | - Alán Aspuru-Guzik
- Chemical Physics Theory Group , Department of Chemistry , University of Toronto , Toronto , Ontario M5S 3H6 , Canada
- Department of Computer Science , University of Toronto , 214 College St. , Toronto , Ontario M5T 3A1 , Canada
- Vector Institute for Artificial Intelligence , 661 University Ave. Suite 710 , Toronto , Ontario M5G 1M1 , Canada
- Lebovic Fellow , Canadian Institute for Advanced Research (CIFAR) , 661 University Ave , Toronto , ON M5G 1M1 , Canada
| | - David Balcells
- Hylleraas Centre for Quantum Molecular Sciences , Department of Chemistry , University of Oslo , P. O. Box 1033, Blindern , N-0315 , Oslo , Norway .
| |
Collapse
|
68
|
Kuroda D, Tsumoto K. Engineering Stability, Viscosity, and Immunogenicity of Antibodies by Computational Design. J Pharm Sci 2020; 109:1631-1651. [DOI: 10.1016/j.xphs.2020.01.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/25/2019] [Accepted: 01/10/2020] [Indexed: 12/18/2022]
|
69
|
Puertas-Martín S, Banegas-Luna AJ, Paredes-Ramos M, Redondo JL, Ortigosa PM, Brovarets' OO, Pérez-Sánchez H. Is high performance computing a requirement for novel drug discovery and how will this impact academic efforts? Expert Opin Drug Discov 2020; 15:981-986. [PMID: 32345062 DOI: 10.1080/17460441.2020.1758664] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Savíns Puertas-Martín
- Supercomputing - Algorithms Research Group (SAL), University of Almería, Agrifood Campus of International Excellence , Almería, Spain
| | - Antonio J Banegas-Luna
- Bioinformatics and High Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Universidad Católica San Antonio De Murcia (UCAM) , Murcia, Spain
| | - María Paredes-Ramos
- METMED Research Group, Physical Chemistry Department, Universidade Da Coruña (UDC) , Coruña, Spain
| | - Juana L Redondo
- Supercomputing - Algorithms Research Group (SAL), University of Almería, Agrifood Campus of International Excellence , Almería, Spain
| | - Pilar M Ortigosa
- Supercomputing - Algorithms Research Group (SAL), University of Almería, Agrifood Campus of International Excellence , Almería, Spain
| | - Ol'ha O Brovarets'
- Department of Molecular and Quantum Biophysics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine , Kyiv, Ukraine
| | - Horacio Pérez-Sánchez
- Bioinformatics and High Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Universidad Católica San Antonio De Murcia (UCAM) , Murcia, Spain
| |
Collapse
|
70
|
Chávez-Hernández AL, Sánchez-Cruz N, Medina-Franco JL. A Fragment Library of Natural Products and its Comparative Chemoinformatic Characterization. Mol Inform 2020; 39:e2000050. [PMID: 32302465 DOI: 10.1002/minf.202000050] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/17/2020] [Indexed: 11/06/2022]
Abstract
We report a comprehensive fragment library with 205,903 fragments derived from the recently published Collection of Open Natural Products (COCONUT) data set with more than 400,000 non-redundant natural products. The natural products-based fragment library was compared with other two fragment libraries herein generated from ChEMBL (biologically relevant compounds) and Enamine-REAL (a large on-demand collection of synthetic compounds), both used as reference data sets with relevance in drug discovery. It was found that there is a large diversity of unique fragments derived from natural products and that the entire structures and fragments derived from natural products are more diverse and structurally complex than the two reference compound collections. During this work we introduced a novel visual representation of the chemical space based on the recently published concept of statistical-based database fingerprint. The compounds and fragments libraries from natural products generated and analyzed in this work are freely available.
Collapse
Affiliation(s)
- Ana L Chávez-Hernández
- Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Mexico City, 04510, Mexico
| | - Norberto Sánchez-Cruz
- Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Mexico City, 04510, Mexico
| | - José L Medina-Franco
- Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Mexico City, 04510, Mexico
| |
Collapse
|
71
|
Grebner C, Matter H, Plowright AT, Hessler G. Automated De Novo Design in Medicinal Chemistry: Which Types of Chemistry Does a Generative Neural Network Learn? J Med Chem 2020; 63:8809-8823. [PMID: 32134646 DOI: 10.1021/acs.jmedchem.9b02044] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Artificial intelligence offers promising solutions for property prediction, compound design, and retrosynthetic planning, which are expected to significantly accelerate the search for pharmacologically relevant molecules. Here, we investigate aspects of artificial intelligence based de novo design pertaining to its integration into real-life workflows. First, different chemical spaces were used as training sets for reinforcement learning (RL) in combination with different reward functions. With the trained neuronal networks different biologically active molecules could be regenerated. Excluding molecules with substructures such as five-membered rings from training spaces nevertheless produced results containing these moieties. Furthermore, different scoring functions in RL were investigated and produced different design ensembles. In summary, some of these design proposals are close in chemical space to the query, thus supporting lead optimization, while 3D-shape or QSAR (quantitative structure-activity relationship) models produced significantly different proposals by sampling a broader region of the chemical space, thus supporting lead generation. Therefore, RL provides a good framework to tailored design approaches for different discovery phases.
Collapse
Affiliation(s)
- Christoph Grebner
- Synthetic Molecular Design, Integrated Drug Discovery, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Hans Matter
- Synthetic Molecular Design, Integrated Drug Discovery, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Alleyn T Plowright
- Synthetic Molecular Design, Integrated Drug Discovery, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Gerhard Hessler
- Synthetic Molecular Design, Integrated Drug Discovery, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| |
Collapse
|
72
|
Singh N, Chaput L, Villoutreix BO. Virtual screening web servers: designing chemical probes and drug candidates in the cyberspace. Brief Bioinform 2020; 22:1790-1818. [PMID: 32187356 PMCID: PMC7986591 DOI: 10.1093/bib/bbaa034] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The interplay between life sciences and advancing technology drives a continuous cycle of chemical data growth; these data are most often stored in open or partially open databases. In parallel, many different types of algorithms are being developed to manipulate these chemical objects and associated bioactivity data. Virtual screening methods are among the most popular computational approaches in pharmaceutical research. Today, user-friendly web-based tools are available to help scientists perform virtual screening experiments. This article provides an overview of internet resources enabling and supporting chemical biology and early drug discovery with a main emphasis on web servers dedicated to virtual ligand screening and small-molecule docking. This survey first introduces some key concepts and then presents recent and easily accessible virtual screening and related target-fishing tools as well as briefly discusses case studies enabled by some of these web services. Notwithstanding further improvements, already available web-based tools not only contribute to the design of bioactive molecules and assist drug repositioning but also help to generate new ideas and explore different hypotheses in a timely fashion while contributing to teaching in the field of drug development.
Collapse
Affiliation(s)
- Natesh Singh
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Ludovic Chaput
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Bruno O Villoutreix
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 Drugs and Molecules for Living Systems, F-59000 Lille, France
| |
Collapse
|
73
|
de Souza Neto LR, Moreira-Filho JT, Neves BJ, Maidana RLBR, Guimarães ACR, Furnham N, Andrade CH, Silva FP. In silico Strategies to Support Fragment-to-Lead Optimization in Drug Discovery. Front Chem 2020; 8:93. [PMID: 32133344 PMCID: PMC7040036 DOI: 10.3389/fchem.2020.00093] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/30/2020] [Indexed: 12/16/2022] Open
Abstract
Fragment-based drug (or lead) discovery (FBDD or FBLD) has developed in the last two decades to become a successful key technology in the pharmaceutical industry for early stage drug discovery and development. The FBDD strategy consists of screening low molecular weight compounds against macromolecular targets (usually proteins) of clinical relevance. These small molecular fragments can bind at one or more sites on the target and act as starting points for the development of lead compounds. In developing the fragments attractive features that can translate into compounds with favorable physical, pharmacokinetics and toxicity (ADMET-absorption, distribution, metabolism, excretion, and toxicity) properties can be integrated. Structure-enabled fragment screening campaigns use a combination of screening by a range of biophysical techniques, such as differential scanning fluorimetry, surface plasmon resonance, and thermophoresis, followed by structural characterization of fragment binding using NMR or X-ray crystallography. Structural characterization is also used in subsequent analysis for growing fragments of selected screening hits. The latest iteration of the FBDD workflow employs a high-throughput methodology of massively parallel screening by X-ray crystallography of individually soaked fragments. In this review we will outline the FBDD strategies and explore a variety of in silico approaches to support the follow-up fragment-to-lead optimization of either: growing, linking, and merging. These fragment expansion strategies include hot spot analysis, druggability prediction, SAR (structure-activity relationships) by catalog methods, application of machine learning/deep learning models for virtual screening and several de novo design methods for proposing synthesizable new compounds. Finally, we will highlight recent case studies in fragment-based drug discovery where in silico methods have successfully contributed to the development of lead compounds.
Collapse
Affiliation(s)
- Lauro Ribeiro de Souza Neto
- LaBECFar – Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - José Teófilo Moreira-Filho
- LabMol – Laboratory for Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Brazil
| | - Bruno Junior Neves
- LabMol – Laboratory for Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Brazil
- Laboratory of Cheminformatics, Centro Universitário de Anápolis – UniEVANGÉLICA, Anápolis, Brazil
| | - Rocío Lucía Beatriz Riveros Maidana
- LaBECFar – Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Laboratório de Genômica Funcional e Bioinformática, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Ana Carolina Ramos Guimarães
- Laboratório de Genômica Funcional e Bioinformática, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Nicholas Furnham
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Carolina Horta Andrade
- LabMol – Laboratory for Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Brazil
| | - Floriano Paes Silva
- LaBECFar – Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
74
|
Affiliation(s)
- Marco Foscato
- Department of Chemistry, University of Bergen, Allégaten 41, N-5007 Bergen, Norway
| | - Vidar R. Jensen
- Department of Chemistry, University of Bergen, Allégaten 41, N-5007 Bergen, Norway
| |
Collapse
|
75
|
Schneider P, Walters WP, Plowright AT, Sieroka N, Listgarten J, Goodnow RA, Fisher J, Jansen JM, Duca JS, Rush TS, Zentgraf M, Hill JE, Krutoholow E, Kohler M, Blaney J, Funatsu K, Luebkemann C, Schneider G. Rethinking drug design in the artificial intelligence era. Nat Rev Drug Discov 2019. [DOI: 78495111110.1038/s41573-019-0050-3' target='_blank'>'"<>78495111110.1038/s41573-019-0050-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [78495111110.1038/s41573-019-0050-3','', '10.1002/anie.201814681')">Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
78495111110.1038/s41573-019-0050-3" />
|
76
|
Rethinking drug design in the artificial intelligence era. Nat Rev Drug Discov 2019; 19:353-364. [PMID: 31801986 DOI: 10.1038/s41573-019-0050-3] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2019] [Indexed: 12/17/2022]
|
77
|
Empel C, Koenigs RM. Künstliche Intelligenz in der organischen Synthese – en route zu autonomer Synthese? Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201911062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Claire Empel
- Institut für Organische ChemieRWTH Aachen Landoltweg 1 52074 Aachen Deutschland
| | - Rene M. Koenigs
- Institut für Organische ChemieRWTH Aachen Landoltweg 1 52074 Aachen Deutschland
| |
Collapse
|
78
|
Zhao Z, Dai X, Li C, Wang X, Tian J, Feng Y, Xie J, Ma C, Nie Z, Fan P, Qian M, He X, Wu S, Zhang Y, Zheng X. Pyrazolone structural motif in medicinal chemistry: Retrospect and prospect. Eur J Med Chem 2019; 186:111893. [PMID: 31761383 PMCID: PMC7115706 DOI: 10.1016/j.ejmech.2019.111893] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 12/13/2022]
Abstract
The pyrazolone structural motif is a critical element of drugs aimed at different biological end-points. Medicinal chemistry researches have synthesized drug-like pyrazolone candidates with several medicinal features including antimicrobial, antitumor, CNS (central nervous system) effect, anti-inflammatory activities and so on. Meanwhile, SAR (Structure-Activity Relationship) investigations have drawn attentions among medicinal chemists, along with a plenty of analogues have been derived for multiple targets. In this review, we comprehensively summarize the biological activity and SAR for pyrazolone analogues, wishing to give an overall retrospect and prospect on the pyrazolone derivatives. The pyrazolone structural motif is a critical element of drugs aimed at different biological end-points. The pyrazolone analogues have been carried out to drug-like candidates with broad range of medicinal properties. This review wishes to give an overall retrospect and prospect on the pyrazolone derivatives.
Collapse
Affiliation(s)
- Zefeng Zhao
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Xufen Dai
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Chenyang Li
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Xiao Wang
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Jiale Tian
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Ying Feng
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Jing Xie
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Cong Ma
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Zhuang Nie
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Peinan Fan
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Mingcheng Qian
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, 213164, Jiangsu, China; Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000, Ghent, Belgium
| | - Xirui He
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Shaoping Wu
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China.
| | - Yongmin Zhang
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China; Sorbonne Université, Institut Parisien de Chimie Moléculaire, CNRS UMR 8232, 4 Place Jussieu, 75005, Paris, France
| | - Xiaohui Zheng
- School of Pharmacy, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| |
Collapse
|
79
|
Empel C, Koenigs RM. Artificial-Intelligence-Driven Organic Synthesis-En Route towards Autonomous Synthesis? Angew Chem Int Ed Engl 2019; 58:17114-17116. [PMID: 31638733 DOI: 10.1002/anie.201911062] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Indexed: 11/11/2022]
Abstract
AI for chemistry: Automated synthesis can now be performed using an artificial intelligence algorithm to propose the synthetic route and a robotic microfluidic platform to execute the synthesis. This Highlight describes this approach towards small-molecule synthesis and reflects on the significance of this milestone in chemistry.
Collapse
Affiliation(s)
- Claire Empel
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074, Aachen, Germany
| | - Rene M Koenigs
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074, Aachen, Germany
| |
Collapse
|
80
|
Chen H, Engkvist O. Has Drug Design Augmented by Artificial Intelligence Become a Reality? Trends Pharmacol Sci 2019; 40:806-809. [PMID: 31629547 DOI: 10.1016/j.tips.2019.09.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 09/18/2019] [Indexed: 11/30/2022]
Abstract
The application of artificial intelligence (AI) to drug discovery has become a hot topic in recent years. Generative molecular design based on deep learning is a particular an area of attention. Zhavoronkov et al. recently published a novel approach in which de novo molecular design based on deep learning was used to discover novel potent DDR1 kinase inhibitors. It took 21 days from model building to compound design, and a total of six AI-designed compounds were synthesized and tested. The study highlights how quickly the field of AI-designed compounds is developing, and we can expect further developments in the coming years.
Collapse
Affiliation(s)
- Hongming Chen
- Hit Discovery, Discovery Sciences, Biopharmaceutics R&D, AstraZeneca, Gothenburg, Sweden.
| | - Ola Engkvist
- Hit Discovery, Discovery Sciences, Biopharmaceutics R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
81
|
Bruns D, Merk D, Santhana Kumar K, Baumgartner M, Schneider G. Synthetic Activators of Cell Migration Designed by Constructive Machine Learning. ChemistryOpen 2019; 8:1303-1308. [PMID: 31660283 PMCID: PMC6807213 DOI: 10.1002/open.201900222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/11/2019] [Indexed: 11/26/2022] Open
Abstract
Constructive machine learning aims to create examples from its learned domain which are likely to exhibit similar properties. Here, a recurrent neural network was trained with the chemical structures of known cell-migration modulators. This machine learning model was used to generate new molecules that mimic the training compounds. Two top-scoring designs were synthesized, and tested for functional activity in a phenotypic spheroid cell migration assay. These computationally generated small molecules significantly increased the migration of medulloblastoma cells. The results further corroborate the applicability of constructive machine learning to the de novo design of druglike molecules with desired properties.
Collapse
Affiliation(s)
- Dominique Bruns
- ETH Zurich, Department ofChemistry and Applied BiosciencesVladimir-Prelog-Weg 4CH-8093ZurichSwitzerland
| | - Daniel Merk
- ETH Zurich, Department ofChemistry and Applied BiosciencesVladimir-Prelog-Weg 4CH-8093ZurichSwitzerland
| | - Karthiga Santhana Kumar
- Pediatric Neuro-OncologyResearch Group, Department of Oncology, Children's Research Center, University Children's Hospital ZurichLengghalde 5CH-8008ZurichSwitzerland
| | - Martin Baumgartner
- Pediatric Neuro-OncologyResearch Group, Department of Oncology, Children's Research Center, University Children's Hospital ZurichLengghalde 5CH-8008ZurichSwitzerland
| | - Gisbert Schneider
- ETH Zurich, Department ofChemistry and Applied BiosciencesVladimir-Prelog-Weg 4CH-8093ZurichSwitzerland
| |
Collapse
|
82
|
Yang X, Wang Y, Byrne R, Schneider G, Yang S. Concepts of Artificial Intelligence for Computer-Assisted Drug Discovery. Chem Rev 2019; 119:10520-10594. [PMID: 31294972 DOI: 10.1021/acs.chemrev.8b00728] [Citation(s) in RCA: 369] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Artificial intelligence (AI), and, in particular, deep learning as a subcategory of AI, provides opportunities for the discovery and development of innovative drugs. Various machine learning approaches have recently (re)emerged, some of which may be considered instances of domain-specific AI which have been successfully employed for drug discovery and design. This review provides a comprehensive portrayal of these machine learning techniques and of their applications in medicinal chemistry. After introducing the basic principles, alongside some application notes, of the various machine learning algorithms, the current state-of-the art of AI-assisted pharmaceutical discovery is discussed, including applications in structure- and ligand-based virtual screening, de novo drug design, physicochemical and pharmacokinetic property prediction, drug repurposing, and related aspects. Finally, several challenges and limitations of the current methods are summarized, with a view to potential future directions for AI-assisted drug discovery and design.
Collapse
Affiliation(s)
- Xin Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| | - Yifei Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| | - Ryan Byrne
- ETH Zurich , Department of Chemistry and Applied Biosciences , Vladimir-Prelog-Weg 4 , CH-8093 Zurich , Switzerland
| | - Gisbert Schneider
- ETH Zurich , Department of Chemistry and Applied Biosciences , Vladimir-Prelog-Weg 4 , CH-8093 Zurich , Switzerland
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| |
Collapse
|
83
|
Abstract
Introduction: The development of drug candidates with a defined selectivity profile and a unique molecular structure is of fundamental interest for drug discovery. In contrast to the costly screening of large substance libraries, the targeted de novo design of a drug by using structural information of either the biological target and/or structure-activity relationship data of active modulators offers an efficient and intellectually appealing alternative. Areas covered: This review provides an overview on the different techniques of de novo drug design (ligand-based drug design, structure-based drug design, and fragment-based drug design) and highlights successful examples of this targeted approach toward selective modulators of therapeutically relevant targets. Expert opinion: De novo drug design has established itself as a very efficient method for the development of potent and selective modulators for a variety of different biological target classes. The ever-growing wealth of structural data on therapeutic targets will certainly further enhance the importance of de novo design for the drug discovery process in the future. However, a consistent use of the terminology of de novo drug design in the scientific literature should be sought.
Collapse
Affiliation(s)
- Thomas Fischer
- a Center of Organic and Medicinal Chemistry, Institute of Chemistry and Biotechnology , Zurich University of Applied Sciences ZHAW , Wädenswil , Switzerland
| | - Silvia Gazzola
- b Dipartimento di Scienza e Alta Tecnologia , Università degli Studi dell'Insubria , Como , Italy
| | - Rainer Riedl
- a Center of Organic and Medicinal Chemistry, Institute of Chemistry and Biotechnology , Zurich University of Applied Sciences ZHAW , Wädenswil , Switzerland
| |
Collapse
|