1
|
Wang S, Liang D, Wang J, Dong K, Zhang Y, Liang H, Xu X, Song T. FraHMT: A Fragment-Oriented Heterogeneous Graph Molecular Generation Model for Target Proteins. J Chem Inf Model 2024; 64:3718-3732. [PMID: 38644797 DOI: 10.1021/acs.jcim.4c00252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The molecular generation task stands as a pivotal step in the domains of computational chemistry and drug discovery, aiming to computationally generate molecular structures for specific properties. In contrast to previous models that focused primarily on SMILES strings or molecular graphs, our model placed a special emphasis on the substructure information on molecules, enabling the model to learn richer chemical rules and structure features from fragments and chemical reaction information on molecules. To accomplish this, we fragmented the molecules to construct heterogeneous graph representations based on atom and fragment information. Then our model mapped the heterogeneous graph data into a latent vector space by using an encoder and employed a self-regressive generative model as a decoder for molecular generation. Additionally, we performed transfer learning on the model using a small set of ligand molecules known to be active against the target protein to generate molecules that bind better to the target protein. Experimental results demonstrate that our model is highly competitive with state-of-the-art models. It can generate valid and diverse molecules with favorable physicochemical properties and drug-likeness. Importantly, they produce novel molecules with high docking scores against the target proteins.
Collapse
Affiliation(s)
- Shuang Wang
- College of Computer Science and Technology, China University of Petroleum, QingDao 266580, China
| | - Dingming Liang
- College of Computer Science and Technology, China University of Petroleum, QingDao 266580, China
| | - Jianmin Wang
- College of Computer Science and Technology, China University of Petroleum, QingDao 266580, China
- The Interdisciplinary Graduate Program in Integrative Biotechnology, Yonsei University, Incheon 21983, Republic of Korea
| | - Kaiyu Dong
- College of Computer Science and Technology, China University of Petroleum, QingDao 266580, China
| | - Yunjing Zhang
- College of Computer Science and Technology, China University of Petroleum, QingDao 266580, China
| | - Huicong Liang
- Marine Biomedical Institute of Qingdao, School of Medicine and Pharmacy, Ocean University of China, QingDao 266580, China
| | - Ximing Xu
- Marine Biomedical Institute of Qingdao, School of Medicine and Pharmacy, Ocean University of China, QingDao 266580, China
| | - Tao Song
- College of Computer Science and Technology, China University of Petroleum, QingDao 266580, China
- Department of Artificial Intelligence, Faculty of Computer Science, Polytechnical University of Madrid, Madrid 28031, Spain
| |
Collapse
|
2
|
Singh K, Bhushan B, Singh B. Advances in Drug Discovery and Design using Computer-aided Molecular Modeling. Curr Comput Aided Drug Des 2024; 20:697-710. [PMID: 37711101 DOI: 10.2174/1573409920666230914123005] [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: 07/04/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 09/16/2023]
Abstract
Computer-aided molecular modeling is a rapidly emerging technology that is being used to accelerate the discovery and design of new drug therapies. It involves the use of computer algorithms and 3D structures of molecules to predict interactions between molecules and their behavior in the body. This has drastically improved the speed and accuracy of drug discovery and design. Additionally, computer-aided molecular modeling has the potential to reduce costs, increase the quality of data, and identify promising targets for drug development. Through the use of sophisticated methods, such as virtual screening, molecular docking, pharmacophore modeling, and quantitative structure-activity relationships, scientists can achieve higher levels of efficacy and safety for new drugs. Moreover, it can be used to understand the activity of known drugs and simplify the process of formulating, optimizing, and predicting the pharmacokinetics of new and existing drugs. In conclusion, computer-aided molecular modeling is an effective tool to rapidly progress drug discovery and design by predicting the interactions between molecules and anticipating the behavior of new drugs in the body.
Collapse
Affiliation(s)
- Kuldeep Singh
- Department of Pharmacology, Rajiv Academy for Pharmacy, Mathura Uttar Pradesh, India
| | - Bharat Bhushan
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura Uttar Pradesh, India
| | - Bhoopendra Singh
- Department of Pharmacy, B.S.A. College of Engineering & Technology, Mathura Uttar Pradesh India
| |
Collapse
|
3
|
Talevi A. Computer-Aided Drug Discovery and Design: Recent Advances and Future Prospects. Methods Mol Biol 2024; 2714:1-20. [PMID: 37676590 DOI: 10.1007/978-1-0716-3441-7_1] [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] [Indexed: 09/08/2023]
Abstract
Computer-aided drug discovery and design involve the use of information technologies to identify and develop, on a rational ground, chemical compounds that align a set of desired physicochemical and biological properties. In its most common form, it involves the identification and/or modification of an active scaffold (or the combination of known active scaffolds), although de novo drug design from scratch is also possible. Traditionally, the drug discovery and design processes have focused on the molecular determinants of the interactions between drug candidates and their known or intended pharmacological target(s). Nevertheless, in modern times, drug discovery and design are conceived as a particularly complex multiparameter optimization task, due to the complicated, often conflicting, property requirements.This chapter provides an updated overview of in silico approaches for identifying active scaffolds and guiding the subsequent optimization process. Recent groundbreaking advances in the field have also analyzed the integration of state-of-the-art machine learning approaches in every step of the drug discovery process (from prediction of target structure to customized molecular docking scoring functions), integration of multilevel omics data, and the use of a diversity of computational approaches to assist target validation and assess plausible binding pockets.
Collapse
Affiliation(s)
- Alan Talevi
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, National University of La Plata (UNLP), La Plata, Argentina.
- Argentinean National Council of Scientific and Technical Research (CONICET), La Plata, Argentina.
| |
Collapse
|
4
|
Amoroso N, Gambacorta N, Mastrolorito F, Togo MV, Trisciuzzi D, Monaco A, Pantaleo E, Altomare CD, Ciriaco F, Nicolotti O. Making sense of chemical space network shows signs of criticality. Sci Rep 2023; 13:21335. [PMID: 38049451 PMCID: PMC10696027 DOI: 10.1038/s41598-023-48107-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/22/2023] [Indexed: 12/06/2023] Open
Abstract
Chemical space modelling has great importance in unveiling and visualising latent information, which is critical in predictive toxicology related to drug discovery process. While the use of traditional molecular descriptors and fingerprints may suffer from the so-called curse of dimensionality, complex networks are devoid of the typical drawbacks of coordinate-based representations. Herein, we use chemical space networks (CSNs) to analyse the case of the developmental toxicity (Dev Tox), which remains a challenging endpoint for the difficulty of gathering enough reliable data despite very important for the protection of the maternal and child health. Our study proved that the Dev Tox CSN has a complex non-random organisation and can thus provide a wealth of meaningful information also for predictive purposes. At a phase transition, chemical similarities highlight well-established toxicophores, such as aryl derivatives, mostly neurotoxic hydantoins, barbiturates and amino alcohols, steroids, and volatile organic compounds ether-like chemicals, which are strongly suspected of the Dev Tox onset and can thus be employed as effective alerts for prioritising chemicals before testing.
Collapse
Affiliation(s)
- Nicola Amoroso
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli studi di Bari Aldo Moro, via E. Orabona, 4, 70125, Bari, Italy.
- Istituto Nazionale di Fisica Nucleare, Sezione di Bari, via E. Orabona, 4, 70125, Bari, Italy.
| | - Nicola Gambacorta
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli studi di Bari Aldo Moro, via E. Orabona, 4, 70125, Bari, Italy
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo (Foggia), Italy
| | - Fabrizio Mastrolorito
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli studi di Bari Aldo Moro, via E. Orabona, 4, 70125, Bari, Italy
| | - Maria Vittoria Togo
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli studi di Bari Aldo Moro, via E. Orabona, 4, 70125, Bari, Italy
| | - Daniela Trisciuzzi
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli studi di Bari Aldo Moro, via E. Orabona, 4, 70125, Bari, Italy
| | - Alfonso Monaco
- Istituto Nazionale di Fisica Nucleare, Sezione di Bari, via E. Orabona, 4, 70125, Bari, Italy
- Dipartimento Interateneo di Fisica "M. Merlin", Università degli studi di Bari Aldo Moro, Via Giovanni Amendola, 173, 70125, Bari, Italy
| | - Ester Pantaleo
- Istituto Nazionale di Fisica Nucleare, Sezione di Bari, via E. Orabona, 4, 70125, Bari, Italy
- Dipartimento Interateneo di Fisica "M. Merlin", Università degli studi di Bari Aldo Moro, Via Giovanni Amendola, 173, 70125, Bari, Italy
| | - Cosimo Damiano Altomare
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli studi di Bari Aldo Moro, via E. Orabona, 4, 70125, Bari, Italy
| | - Fulvio Ciriaco
- Dipartimento di Chimica, Università degli studi di Bari Aldo Moro, via E. Orabona, 4, 70125, Bari, Italy.
| | - Orazio Nicolotti
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli studi di Bari Aldo Moro, via E. Orabona, 4, 70125, Bari, Italy
| |
Collapse
|
5
|
Sivangi KB, Amilpur S, Dasari CM. ReGen-DTI: A novel generative drug target interaction model for predicting potential drug candidates against SARS-COV2. Comput Biol Chem 2023; 106:107927. [PMID: 37499436 DOI: 10.1016/j.compbiolchem.2023.107927] [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: 05/16/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/29/2023]
Abstract
Covid-19 has caused massive numbers of infections and fatalities globally. In response, there has been a large-scale experimental and computational research effort to study and develop drugs. Towards this, Deep learning techniques are used for the generation of potential novel drug candidates that are proven to be effective against exploring large molecular search spaces. Recent advances in reinforcement learning in conjunction with generative techniques has proven to be a promising field in the area of drug discovery. In this regard, we propose a generative drug discovery approach using reinforcement techniques for sampling novel molecules that bind to the main protease of SARS-COV2. The generative method reported significant validity scores for the generated novel molecules and captured the underlying features of the training molecules. Further, the model is fine-tuned on existing re-purposed molecules which are active towards specific target proteins based on similarity metrics. Upon fine tuning the model generated 92.71% valid, 93.55% unique, and 100% novel molecules. Unlike previous methods which are dependent on docking procedures, we proposed a deep learning based novel drug target interaction (DTI) model to find the binding affinity between candidate molecules and target protease sequence. Finally, the binding affinity of the generated molecules is predicted against the 3CLPro main protease by using the proposed DTI model. Most of the generated molecules have shown binding affinity scores <100 nM (lower the better), which are significantly better compared to the existing commercial drugs including Remdesevir.
Collapse
Affiliation(s)
- Kaushik Bhargav Sivangi
- Indian Institute of Information Technology, Sri City, Chittoor, 517646, Andhra Pradesh, India
| | - Santhosh Amilpur
- Indian Institute of Information Technology, Sri City, Chittoor, 517646, Andhra Pradesh, India
| | - Chandra Mohan Dasari
- Indian Institute of Information Technology, Sri City, Chittoor, 517646, Andhra Pradesh, India.
| |
Collapse
|
6
|
Schaefer D, Cheng X. Recent Advances in Covalent Drug Discovery. Pharmaceuticals (Basel) 2023; 16:ph16050663. [PMID: 37242447 DOI: 10.3390/ph16050663] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/10/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
In spite of the increasing number of biologics license applications, the development of covalent inhibitors is still a growing field within drug discovery. The successful approval of some covalent protein kinase inhibitors, such as ibrutinib (BTK covalent inhibitor) and dacomitinib (EGFR covalent inhibitor), and the very recent discovery of covalent inhibitors for viral proteases, such as boceprevir, narlaprevir, and nirmatrelvir, represent a new milestone in covalent drug development. Generally, the formation of covalent bonds that target proteins can offer drugs diverse advantages in terms of target selectivity, drug resistance, and administration concentration. The most important factor for covalent inhibitors is the electrophile (warhead), which dictates selectivity, reactivity, and the type of protein binding (i.e., reversible or irreversible) and can be modified/optimized through rational designs. Furthermore, covalent inhibitors are becoming more and more common in proteolysis, targeting chimeras (PROTACs) for degrading proteins, including those that are currently considered to be 'undruggable'. The aim of this review is to highlight the current state of covalent inhibitor development, including a short historical overview and some examples of applications of PROTAC technologies and treatment of the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Daniel Schaefer
- Buchmann Institute for Molecular Life Sciences, Chemical Biology, Goethe University Frankfurt am Main, Max-von-Laue-Strasse 15. R. 3.652, 60438 Frankfurt am Main, Germany
- Pharmaceutical Chemistry, Goethe University Frankfurt am Main, 60438 Frankfurt am Main, Germany
| | - Xinlai Cheng
- Buchmann Institute for Molecular Life Sciences, Chemical Biology, Goethe University Frankfurt am Main, Max-von-Laue-Strasse 15. R. 3.652, 60438 Frankfurt am Main, Germany
- Pharmaceutical Chemistry, Goethe University Frankfurt am Main, 60438 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, 60596 Frankfurt am Main, Germany
| |
Collapse
|
7
|
De Novo Design of Imidazopyridine-Tethered Pyrazolines That Target Phosphorylation of STAT3 in Human Breast Cancer Cells. Bioengineering (Basel) 2023; 10:bioengineering10020159. [PMID: 36829653 PMCID: PMC9952374 DOI: 10.3390/bioengineering10020159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/02/2023] [Accepted: 01/08/2023] [Indexed: 01/26/2023] Open
Abstract
In breast cancer (BC), STAT3 is hyperactivated. This study explored the design of imidazopyridine-tethered pyrazolines as a de novo drug strategy for inhibiting STAT3 phosphorylation in human BC cells. This involved the synthesis and characterization of two series of compounds namely, 1-(3-(2,6-dimethylimidazo [1,2-a]pyridin-3-yl)-5-(3-nitrophenyl)-4,5-dihydro-1H-pyrazol-1-yl)-2-(4-(substituted)piperazin-1-yl)ethanone and N-substituted-3-(2,6-dimethylimidazo[1,2-a]pyridin-3-yl)-5-(3-nitrophenyl)-4,5-dihydro-1H-pyrazoline-1-carbothioamides. Compound 3f with 2,3-dichlorophenyl substitution was recognized among the tested series as a lead structure that inhibited the viability of MCF-7 cells with an IC50 value of 9.2 μM. A dose- and time-dependent inhibition of STAT3 phosphorylation at Tyr705 and Ser727 was observed in MCF-7 and T47D cells when compound 3f was added in vitro. Calculations using density functional theory showed that the title compounds HOMOs and LUMOs are situated on imidazopyridine-pyrazoline and nitrophenyl rings, respectively. Hence, compound 3f effectively inhibited STAT3 phosphorylation in MCF-7 and T47D cells, indicating that these structures may be an alternative synthon to target STAT3 signaling in BC.
Collapse
|
8
|
Sarkar C, Das B, Rawat VS, Wahlang JB, Nongpiur A, Tiewsoh I, Lyngdoh NM, Das D, Bidarolli M, Sony HT. Artificial Intelligence and Machine Learning Technology Driven Modern Drug Discovery and Development. Int J Mol Sci 2023; 24:ijms24032026. [PMID: 36768346 PMCID: PMC9916967 DOI: 10.3390/ijms24032026] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/22/2023] Open
Abstract
The discovery and advances of medicines may be considered as the ultimate relevant translational science effort that adds to human invulnerability and happiness. But advancing a fresh medication is a quite convoluted, costly, and protracted operation, normally costing USD ~2.6 billion and consuming a mean time span of 12 years. Methods to cut back expenditure and hasten new drug discovery have prompted an arduous and compelling brainstorming exercise in the pharmaceutical industry. The engagement of Artificial Intelligence (AI), including the deep-learning (DL) component in particular, has been facilitated by the employment of classified big data, in concert with strikingly reinforced computing prowess and cloud storage, across all fields. AI has energized computer-facilitated drug discovery. An unrestricted espousing of machine learning (ML), especially DL, in many scientific specialties, and the technological refinements in computing hardware and software, in concert with various aspects of the problem, sustain this progress. ML algorithms have been extensively engaged for computer-facilitated drug discovery. DL methods, such as artificial neural networks (ANNs) comprising multiple buried processing layers, have of late seen a resurgence due to their capability to power automatic attribute elicitations from the input data, coupled with their ability to obtain nonlinear input-output pertinencies. Such features of DL methods augment classical ML techniques which bank on human-contrived molecular descriptors. A major part of the early reluctance concerning utility of AI in pharmaceutical discovery has begun to melt, thereby advancing medicinal chemistry. AI, along with modern experimental technical knowledge, is anticipated to invigorate the quest for new and improved pharmaceuticals in an expeditious, economical, and increasingly compelling manner. DL-facilitated methods have just initiated kickstarting for some integral issues in drug discovery. Many technological advances, such as "message-passing paradigms", "spatial-symmetry-preserving networks", "hybrid de novo design", and other ingenious ML exemplars, will definitely come to be pervasively widespread and help dissect many of the biggest, and most intriguing inquiries. Open data allocation and model augmentation will exert a decisive hold during the progress of drug discovery employing AI. This review will address the impending utilizations of AI to refine and bolster the drug discovery operation.
Collapse
Affiliation(s)
- Chayna Sarkar
- Department of Pharmacology, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences (NEIGRIHMS), Mawdiangdiang, Shillong 793018, Meghalaya, India
| | - Biswadeep Das
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India
- Correspondence: ; Tel./Fax: +91-135-708-856-0009
| | - Vikram Singh Rawat
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India
| | - Julie Birdie Wahlang
- Department of Pharmacology, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences (NEIGRIHMS), Mawdiangdiang, Shillong 793018, Meghalaya, India
| | - Arvind Nongpiur
- Department of Psychiatry, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences (NEIGRIHMS), Mawdiangdiang, Shillong 793018, Meghalaya, India
| | - Iadarilang Tiewsoh
- Department of Medicine, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences (NEIGRIHMS), Mawdiangdiang, Shillong 793018, Meghalaya, India
| | - Nari M. Lyngdoh
- Department of Anesthesiology, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences (NEIGRIHMS), Mawdiangdiang, Shillong 793018, Meghalaya, India
| | - Debasmita Das
- Department of Computer Science and Engineering, Vellore Institute of Technology, Vellore Campus, Tiruvalam Road, Katpadi, Vellore 632014, Tamil Nadu, India
| | - Manjunath Bidarolli
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India
| | - Hannah Theresa Sony
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India
| |
Collapse
|
9
|
Furka Á. Forty years of combinatorial technology. Drug Discov Today 2022; 27:103308. [PMID: 35760283 DOI: 10.1016/j.drudis.2022.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/24/2022] [Accepted: 06/21/2022] [Indexed: 12/15/2022]
Abstract
Combinatorial technology has been facilitating the synthesis and screening of large molecular libraries containing millions of organic compounds ever since its introduction 40 years ago. It has changed the paradigms of pharmaceutical research from focusing on single compounds to focusing on immense collections of compounds. It inspired the development of dynamic combinatorial libraries, fragment-based drug discovery and virtual library screening. Combinatorial technology was revitalized by the development of DNA encoding. Amplification of DNA oligomers plus next-generation sequencing has made it possible to successfully screen billions of compounds in a single process.
Collapse
Affiliation(s)
- Árpád Furka
- Eötvös Loránd University Budapest Hungary, 1077 Rozsa u. 23-25, Budapest, Hungary.
| |
Collapse
|
10
|
Kulczyk S, Koszytkowska-Stawińska M. Novel drug design framework as a response to neglected and emerging diseases. J Biomol Struct Dyn 2022:1-12. [DOI: 10.1080/07391102.2022.2110519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Stanisław Kulczyk
- Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | | |
Collapse
|
11
|
Lu C, Liu S, Shi W, Yu J, Zhou Z, Zhang X, Lu X, Cai F, Xia N, Wang Y. Systemic evolutionary chemical space exploration for drug discovery. J Cheminform 2022; 14:19. [PMID: 35365231 PMCID: PMC8973791 DOI: 10.1186/s13321-022-00598-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/11/2022] [Indexed: 11/29/2022] Open
Abstract
Chemical space exploration is a major task of the hit-finding process during the pursuit of novel chemical entities. Compared with other screening technologies, computational de novo design has become a popular approach to overcome the limitation of current chemical libraries. Here, we reported a de novo design platform named systemic evolutionary chemical space explorer (SECSE). The platform was conceptually inspired by fragment-based drug design, that miniaturized a “lego-building” process within the pocket of a certain target. The key to virtual hits generation was then turned into a computational search problem. To enhance search and optimization, human intelligence and deep learning were integrated. Application of SECSE against phosphoglycerate dehydrogenase (PHGDH), proved its potential in finding novel and diverse small molecules that are attractive starting points for further validation. This platform is open-sourced and the code is available at http://github.com/KeenThera/SECSE.
Collapse
Affiliation(s)
- Chong Lu
- Keen Therapeutics Co., Ltd., Shanghai, China
| | - Shien Liu
- Keen Therapeutics Co., Ltd., Shanghai, China
| | - Weihua Shi
- Keen Therapeutics Co., Ltd., Shanghai, China
| | - Jun Yu
- Keen Therapeutics Co., Ltd., Shanghai, China
| | - Zhou Zhou
- Keen Therapeutics Co., Ltd., Shanghai, China
| | | | - Xiaoli Lu
- Keen Therapeutics Co., Ltd., Shanghai, China
| | - Faji Cai
- Keen Therapeutics Co., Ltd., Shanghai, China
| | | | - Yikai Wang
- Keen Therapeutics Co., Ltd., Shanghai, China.
| |
Collapse
|
12
|
Shulga DA, Ivanov NN, Palyulin VA. In Silico Structure-Based Approach for Group Efficiency Estimation in Fragment-Based Drug Design Using Evaluation of Fragment Contributions. Molecules 2022; 27:1985. [PMID: 35335347 PMCID: PMC8951103 DOI: 10.3390/molecules27061985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/10/2022] [Accepted: 03/15/2022] [Indexed: 12/10/2022] Open
Abstract
The notion of a contribution of a specific group in an organic molecule's property and/or activity is both common in our thinking and is still not strictly correct due to the inherent non-additivity of free energy with respect to molecular fragments composing a molecule. The fragment- based drug discovery (FBDD) approach has proven to be fruitful in addressing the above notions. The main difficulty of the FBDD, however, is in its reliance on the low throughput and expensive experimental means of determining the fragment-sized molecules binding. In this article we propose a way to enhance the throughput and availability of the FBDD methods by judiciously using an in silico means of assessing the contribution to ligand-receptor binding energy of fragments of a molecule under question using a previously developed in silico Reverse Fragment Based Drug Discovery (R-FBDD) approach. It has been shown that the proposed structure-based drug discovery (SBDD) type of approach fills in the vacant niche among the existing in silico approaches, which mainly stem from the ligand-based drug discovery (LBDD) counterparts. In order to illustrate the applicability of the approach, our work retrospectively repeats the findings of the use case of an FBDD hit-to-lead project devoted to the experimentally based determination of additive group efficiency (GE)-an analog of ligand efficiency (LE) for a group in the molecule-using the Free-Wilson (FW) decomposition. It is shown that in using our in silico approach to evaluate fragment contributions of a ligand and to estimate GE one can arrive at similar decisions as those made using the experimentally determined activity-based FW decomposition. It is also shown that the approach is rather robust to the choice of the scoring function, provided the latter demonstrates a decent scoring power. We argue that the proposed approach of in silico assessment of GE has a wider applicability domain and expect that it will be widely applicable to enhance the net throughput of drug discovery based on the FBDD paradigm.
Collapse
Affiliation(s)
- Dmitry A. Shulga
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | | | - Vladimir A. Palyulin
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia;
| |
Collapse
|
13
|
Wu B, Li L, Cui Y, Zheng K. Cross-Adversarial Learning for Molecular Generation in Drug Design. Front Pharmacol 2022; 12:827606. [PMID: 35126153 PMCID: PMC8815768 DOI: 10.3389/fphar.2021.827606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/30/2021] [Indexed: 11/18/2022] Open
Abstract
Molecular generation is an important but challenging task in drug design, as it requires optimization of chemical compound structures as well as many complex properties. Most of the existing methods use deep learning models to generate molecular representations. However, these methods are faced with the problems of generation validity and semantic information of labels. Considering these challenges, we propose a cross-adversarial learning method for molecular generation, CRAG for short, which integrates both the facticity of VAE-based methods and the diversity of GAN-based methods to further exploit the complex properties of Molecules. To be specific, an adversarially regularized encoder-decoder is used to transform molecules from simplified molecular input linear entry specification (SMILES) into discrete variables. Then, the discrete variables are trained to predict property and generate adversarial samples through projected gradient descent with corresponding labels. Our CRAG is trained using an adversarial pattern. Extensive experiments on two widely used benchmarks have demonstrated the effectiveness of our proposed method on a wide spectrum of metrics. We also utilize a novel metric named Novel/Sample to measure the overall generation effectiveness of models. Therefore, CRAG is promising for AI-based molecular design in various chemical applications.
Collapse
Affiliation(s)
- Banghua Wu
- School of Cyber Science and Engineering, Sichuan University, Chengdu, China
| | - Linjie Li
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Yue Cui
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Kai Zheng
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Kai Zheng,
| |
Collapse
|
14
|
Wang S, Song T, Zhang S, Jiang M, Wei Z, Li Z. Molecular substructure tree generative model for de novo drug design. Brief Bioinform 2022; 23:6510156. [PMID: 35039853 DOI: 10.1093/bib/bbab592] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/19/2021] [Accepted: 12/19/2021] [Indexed: 01/19/2023] Open
Abstract
Deep learning shortens the cycle of the drug discovery for its success in extracting features of molecules and proteins. Generating new molecules with deep learning methods could enlarge the molecule space and obtain molecules with specific properties. However, it is also a challenging task considering that the connections between atoms are constrained by chemical rules. Aiming at generating and optimizing new valid molecules, this article proposed Molecular Substructure Tree Generative Model, in which the molecule is generated by adding substructure gradually. The proposed model is based on the Variational Auto-Encoder architecture, which uses the encoder to map molecules to the latent vector space, and then builds an autoregressive generative model as a decoder to generate new molecules from Gaussian distribution. At the same time, for the molecular optimization task, a molecular optimization model based on CycleGAN was constructed. Experiments showed that the model could generate valid and novel molecules, and the optimized model effectively improves the molecular properties.
Collapse
Affiliation(s)
- Shuang Wang
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao 266580, China
| | - Tao Song
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao 266580, China
| | - Shugang Zhang
- College of Computer Science and Technology, Ocean University of China, Qingdao 266100, China
| | - Mingjian Jiang
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao 266033, China
| | - Zhiqiang Wei
- College of Computer Science and Technology, Ocean University of China, Qingdao 266100, China
| | - Zhen Li
- College of Computer Science and Technology, Qingdao University, Qingdao 266071, China
| |
Collapse
|
15
|
Abstract
Abstract
Machine learning (ML) has revolutionised the field of structure-based drug design (SBDD) in recent years. During the training stage, ML techniques typically analyse large amounts of experimentally determined data to create predictive models in order to inform the drug discovery process. Deep learning (DL) is a subfield of ML, that relies on multiple layers of a neural network to extract significantly more complex patterns from experimental data, and has recently become a popular choice in SBDD. This review provides a thorough summary of the recent DL trends in SBDD with a particular focus on de novo drug design, binding site prediction, and binding affinity prediction of small molecules.
Collapse
|
16
|
Amilpur S, Bhukya R. Predicting novel drug candidates against Covid-19 using generative deep neural networks. J Mol Graph Model 2022; 110:108045. [PMID: 34688160 PMCID: PMC8510991 DOI: 10.1016/j.jmgm.2021.108045] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/17/2021] [Accepted: 10/04/2021] [Indexed: 12/21/2022]
Abstract
The novel Coronavirus outbreak has created a massive economic crisis, and many succumb to death, disturbing the lives of mankind all over the world. Currently, there are no viable treatment for this condition, drug development approaches are being pursued with vigor. The major treatment options are to repurpose existing drugs or to find new ones. Traditional methods for drug discovery take a longer time, so there is an urgent need to develop some alternative techniques that reduces search space for drug candidates. Towards this endeavor, we propose a novel drug discovery method that leverages on long short term memory (LSTM) model to generate novel molecules that are adept at binding with novel Coronavirus protease. Our study demonstrates that the proposed method is able to recreate novel molecules that correlate very much with the properties of trained molecules. Further, we fine-tune the model to generate novel drug-like molecules that are active towards a specific target. We consider 3CLPro, the main protease of novel Coronavirus, as a therapeutic target and demonstrated in silico screening to assess target structural binding affinities with docking simulations. We observed that 80% of generated molecules show docking free energy of less than -5.8 kcal/mol. The top generated drug candidate has the highest binding affinity with a docking score of -8.5 kcal/mol, which is very much lower when compared to approved existing commercial drugs including, Remdesivir. The low binding energy indicates that the generated molecules could be explored as potential drug candidates for Covid-19.
Collapse
Affiliation(s)
| | - Raju Bhukya
- National Institute of Technology, Waranagl, 506004, India
| |
Collapse
|
17
|
Drug Discovery for Mycobacterium tuberculosis Using Structure-Based Computer-Aided Drug Design Approach. Int J Mol Sci 2021; 22:ijms222413259. [PMID: 34948055 PMCID: PMC8703488 DOI: 10.3390/ijms222413259] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/09/2021] [Accepted: 11/14/2021] [Indexed: 12/12/2022] Open
Abstract
Developing new, more effective antibiotics against resistant Mycobacterium tuberculosis that inhibit its essential proteins is an appealing strategy for combating the global tuberculosis (TB) epidemic. Finding a compound that can target a particular cavity in a protein and interrupt its enzymatic activity is the crucial objective of drug design and discovery. Such a compound is then subjected to different tests, including clinical trials, to study its effectiveness against the pathogen in the host. In recent times, new techniques, which involve computational and analytical methods, enhanced the chances of drug development, as opposed to traditional drug design methods, which are laborious and time-consuming. The computational techniques in drug design have been improved with a new generation of software used to develop and optimize active compounds that can be used in future chemotherapeutic development to combat global tuberculosis resistance. This review provides an overview of the evolution of tuberculosis resistance, existing drug management, and the design of new anti-tuberculosis drugs developed based on the contributions of computational techniques. Also, we show an appraisal of available software and databases on computational drug design with an insight into the application of this software and databases in the development of anti-tubercular drugs. The review features a perspective involving machine learning, artificial intelligence, quantum computing, and CRISPR combination with available computational techniques as a prospective pathway to design new anti-tubercular drugs to combat resistant tuberculosis.
Collapse
|
18
|
Grebner C, Matter H, Hessler G. Artificial Intelligence in Compound Design. Methods Mol Biol 2021; 2390:349-382. [PMID: 34731477 DOI: 10.1007/978-1-0716-1787-8_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Artificial intelligence has seen an incredibly fast development in recent years. Many novel technologies for property prediction of drug molecules as well as for the design of novel molecules were introduced by different research groups. These artificial intelligence-based design methods can be applied for suggesting novel chemical motifs in lead generation or scaffold hopping as well as for optimization of desired property profiles during lead optimization. In lead generation, broad sampling of the chemical space for identification of novel motifs is required, while in the lead optimization phase, a detailed exploration of the chemical neighborhood of a current lead series is advantageous. These different requirements for successful design outcomes render different combinations of artificial intelligence technologies useful. Overall, we observe that a combination of different approaches with tailored scoring and evaluation schemes appears beneficial for efficient artificial intelligence-based compound design.
Collapse
Affiliation(s)
- Christoph Grebner
- Sanofi-Aventis Deutschland GmbH, R&D, Integrated Drug Discovery, Frankfurt am Main, Germany
| | - Hans Matter
- Sanofi-Aventis Deutschland GmbH, R&D, Integrated Drug Discovery, Frankfurt am Main, Germany
| | - Gerhard Hessler
- Sanofi-Aventis Deutschland GmbH, R&D, Integrated Drug Discovery, Frankfurt am Main, Germany.
| |
Collapse
|
19
|
Chávez-Hernández AL, Juárez-Mercado KE, Saldívar-González FI, Medina-Franco JL. Towards the De Novo Design of HIV-1 Protease Inhibitors Based on Natural Products. Biomolecules 2021; 11:1805. [PMID: 34944448 PMCID: PMC8698858 DOI: 10.3390/biom11121805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/22/2021] [Accepted: 11/29/2021] [Indexed: 01/14/2023] Open
Abstract
Acquired immunodeficiency syndrome (AIDS) caused by the human immunodeficiency virus (HIV) continues to be a public health problem. In 2020, 680,000 people died from HIV-related causes, and 1.5 million people were infected. Antiretrovirals are a way to control HIV infection but not to cure AIDS. As such, effective treatment must be developed to control AIDS. Developing a drug is not an easy task, and there is an enormous amount of work and economic resources invested. For this reason, it is highly convenient to employ computer-aided drug design methods, which can help generate and identify novel molecules. Using the de novo design, novel molecules can be developed using fragments as building blocks. In this work, we develop a virtual focused compound library of HIV-1 viral protease inhibitors from natural product fragments. Natural products are characterized by a large diversity of functional groups, many sp3 atoms, and chiral centers. Pseudo-natural products are a combination of natural products fragments that keep the desired structural characteristics from different natural products. An interactive version of chemical space visualization of virtual compounds focused on HIV-1 viral protease inhibitors from natural product fragments is freely available in the supplementary material.
Collapse
Affiliation(s)
| | | | | | - José L. Medina-Franco
- DIFACQUIM Research Group, Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Mexico City 04510, Mexico; (A.L.C.-H.); (K.E.J.-M.); (F.I.S.-G.)
| |
Collapse
|
20
|
Machine Learning Applied to the Modeling of Pharmacological and ADMET Endpoints. Methods Mol Biol 2021. [PMID: 34731464 DOI: 10.1007/978-1-0716-1787-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
The well-known concept of quantitative structure-activity relationships (QSAR) has been gaining significant interest in the recent years. Data, descriptors, and algorithms are the main pillars to build useful models that support more efficient drug discovery processes with in silico methods. Significant advances in all three areas are the reason for the regained interest in these models. In this book chapter we review various machine learning (ML) approaches that make use of measured in vitro/in vivo data of many compounds. We put these in context with other digital drug discovery methods and present some application examples.
Collapse
|
21
|
Abstract
This review provides the feasible literature on drug discovery through ML tools and techniques that are enforced in every phase of drug development to accelerate the research process and deduce the risk and expenditure in clinical trials. Machine learning techniques improve the decision-making in pharmaceutical data across various applications like QSAR analysis, hit discoveries, de novo drug architectures to retrieve accurate outcomes. Target validation, prognostic biomarkers, digital pathology are considered under problem statements in this review. ML challenges must be applicable for the main cause of inadequacy in interpretability outcomes that may restrict the applications in drug discovery. In clinical trials, absolute and methodological data must be generated to tackle many puzzles in validating ML techniques, improving decision-making, promoting awareness in ML approaches, and deducing risk failures in drug discovery.
Collapse
Affiliation(s)
- Suresh Dara
- Department of Computer Science and Engineering, B V Raju Institute of Technology, Narsapur, Medak, 502313 Telangana India
| | - Swetha Dhamercherla
- Department of Computer Science and Engineering, B V Raju Institute of Technology, Narsapur, Medak, 502313 Telangana India
| | - Surender Singh Jadav
- Centre for Molecular Cancer Research (CMCR) and Vishnu Institute of Pharmaceutical Education and Research (VIPER), Narsapur, Medak, 502313 Telangana India
| | - CH Madhu Babu
- Department of Computer Science and Engineering, B V Raju Institute of Technology, Narsapur, Medak, 502313 Telangana India
| | - Mohamed Jawed Ahsan
- Department of Pharmaceutical Chemistry, Maharishi Arvind College of Pharmacy, Jaipur, 302023 Rajasthan India
| |
Collapse
|
22
|
Piroozmand F, Mohammadipanah F, Sajedi H. Spectrum of deep learning algorithms in drug discovery. Chem Biol Drug Des 2021; 96:886-901. [PMID: 33058458 DOI: 10.1111/cbdd.13674] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022]
Abstract
Deep learning (DL) algorithms are a subset of machine learning algorithms with the aim of modeling complex mapping between a set of elements and their classes. In parallel to the advance in revealing the molecular bases of diseases, a notable innovation has been undertaken to apply DL in data/libraries management, reaction optimizations, differentiating uncertainties, molecule constructions, creating metrics from qualitative results, and prediction of structures or interactions. From source identification to lead discovery and medicinal chemistry of the drug candidate, drug delivery, and modification, the challenges can be subjected to artificial intelligence algorithms to aid in the generation and interpretation of data. Discovery and design approach, both demand automation, large data management and data fusion by the advance in high-throughput mode. The application of DL can accelerate the exploration of drug mechanisms, finding novel indications for existing drugs (drug repositioning), drug development, and preclinical and clinical studies. The impact of DL in the workflow of drug discovery, design, and their complementary tools are highlighted in this review. Additionally, the type of DL algorithms used for this purpose, and their pros and cons along with the dominant directions of future research are presented.
Collapse
Affiliation(s)
- Firoozeh Piroozmand
- Pharmaceutical Biotechnology Lab, Department of Microbiology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Fatemeh Mohammadipanah
- Pharmaceutical Biotechnology Lab, Department of Microbiology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Hedieh Sajedi
- Department of Computer Science, School of Mathematics, Statistics and Computer Science, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
23
|
Jin H, Xia J, Liu Z, Wang XS, Zhang L. A unique ligand-steered strategy for CC chemokine receptor 2 homology modeling to facilitate structure-based virtual screening. Chem Biol Drug Des 2021; 97:944-961. [PMID: 33386704 PMCID: PMC8048943 DOI: 10.1111/cbdd.13820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/12/2020] [Accepted: 12/13/2020] [Indexed: 12/29/2022]
Abstract
CC chemokine receptor 2 (CCR2) antagonists that disrupt CCR2/MCP-1 interaction are expected to treat a variety of inflammatory and autoimmune diseases. The lack of CCR2 crystal structure limits the application of structure-based drug design (SBDD) to this target. Although a few three-dimensional theoretical models have been reported, their accuracy remains to be improved in terms of templates and modeling approaches. In this study, we developed a unique ligand-steered strategy for CCR2 homology modeling. It starts with an initial model based on the X-ray structure of the closest homolog so far, that is, CXCR4. Then, it uses Elastic Network Normal Mode Analysis (EN-NMA) and flexible docking (FD) by AutoDock Vina software to generate ligand-induced fit models. It selects optimal model(s) as well as scoring function(s) via extensive evaluation of model performance based on a unique benchmarking set constructed by our in-house tool, that is, MUBD-DecoyMaker. The model of 81_04 presents the optimal enrichment when combined with the scoring function of PMF04, and the proposed binding mode between CCR2 and Teijin lead by this model complies with the reported mutagenesis data. To highlight the advantage of our strategy, we compared it with the only reported ligand-steered strategy for CCR2 homology modeling, that is, Discovery Studio/Ligand Minimization. Lastly, we performed prospective virtual screening based on 81_04 and CCR2 antagonist bioassay. The identification of two hit compounds, that is, E859-1281 and MolPort-007-767-945, validated the efficacy of our model and the ligand-steered strategy.
Collapse
Affiliation(s)
- Hongwei Jin
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Jie Xia
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesDepartment of New Drug Research and DevelopmentInstitute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Xiang Simon Wang
- Molecular Modeling and Drug Discovery Core for District of Columbia Center for AIDS Research (DC CFAR)Laboratory of Cheminformatics and Drug DesignDepartment of Pharmaceutical SciencesCollege of PharmacyHoward UniversityWashingtonDCUSA
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijingChina
| |
Collapse
|
24
|
In Silico Drug Repurposing by Structural Alteration after Induced Fit: Discovery of a Candidate Agent for Recovery of Nucleotide Excision Repair in Xeroderma Pigmentosum Group D Mutant (R683W). Biomedicines 2021; 9:biomedicines9030249. [PMID: 33802476 PMCID: PMC7999925 DOI: 10.3390/biomedicines9030249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 11/16/2022] Open
Abstract
Xeroderma pigmentosum complementation group D (XPD) is a UV-sensitive syndrome and a rare incurable genetic disease which is caused by the genetic mutation of the excision repair cross-complementation group 2 gene (ERCC2). Patients who harbor only XPD R683W mutant protein develop severe photosensitivity and progressive neurological symptoms. Cultured cells derived from patients with XPD (XPD R683W cells) demonstrate a reduced nucleotide excision repair (NER) ability. We hope to ameliorate clinical symptoms if we can identify candidate agents that would aid recovery of the cells' NER ability. To investigate such candidates, we created in silico methods of drug repurposing (in silico DR), a strategy that utilizes the recovery of ATP-binding in the XPD R683W protein after the induced fit. We chose 4E1RCat and aprepitant as the candidates for our in silico DR, and evaluated them by using the UV-induced unscheduled DNA synthesis (UDS) assay to verify the recovery of NER in XPD R683W cells. UDS values of the cells improved about 1.4-1.7 times after 4E1RCat treatment compared with solvent-only controls; aprepitant showed no positive effect. In this study, therefore, we succeeded in finding the candidate agent 4E1RCat for XPD R683W. We also demonstrated that our in silico DR method is a cost-effective approach for drug candidate discovery.
Collapse
|
25
|
Khan SR, Al Rijjal D, Piro A, Wheeler MB. Integration of AI and traditional medicine in drug discovery. Drug Discov Today 2021; 26:982-992. [PMID: 33476566 DOI: 10.1016/j.drudis.2021.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/01/2020] [Accepted: 01/11/2021] [Indexed: 11/24/2022]
Abstract
AI integration in plant-based traditional medicine could be used to overcome drug discovery challenges.
Collapse
Affiliation(s)
- Saifur R Khan
- Endocrine and Diabetes Platform, Department of Physiology, University of Toronto, Medical Sciences Building, Room 3352, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Advanced Diagnostics, Metabolism, Toronto General Hospital Research Institute, Toronto, ON, Canada.
| | - Dana Al Rijjal
- Endocrine and Diabetes Platform, Department of Physiology, University of Toronto, Medical Sciences Building, Room 3352, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Advanced Diagnostics, Metabolism, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Anthony Piro
- Endocrine and Diabetes Platform, Department of Physiology, University of Toronto, Medical Sciences Building, Room 3352, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Advanced Diagnostics, Metabolism, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Michael B Wheeler
- Endocrine and Diabetes Platform, Department of Physiology, University of Toronto, Medical Sciences Building, Room 3352, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Advanced Diagnostics, Metabolism, Toronto General Hospital Research Institute, Toronto, ON, Canada
| |
Collapse
|
26
|
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
|
27
|
Zhu Y, Che C, Jin B, Zhang N, Su C, Wang F. Knowledge-driven drug repurposing using a comprehensive drug knowledge graph. Health Informatics J 2020; 26:2737-2750. [DOI: 10.1177/1460458220937101] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Due to the huge costs associated with new drug discovery and development, drug repurposing has become an important complement to the traditional de novo approach. With the increasing number of public databases and the rapid development of analytical methodologies, computational approaches have gained great momentum in the field of drug repurposing. In this study, we introduce an approach to knowledge-driven drug repurposing based on a comprehensive drug knowledge graph. We design and develop a drug knowledge graph by systematically integrating multiple drug knowledge bases. We describe path- and embedding-based data representation methods of transforming information in the drug knowledge graph into valuable inputs to allow machine learning models to predict drug repurposing candidates. The evaluation demonstrates that the knowledge-driven approach can produce high predictive results for known diabetes mellitus treatments by only using treatment information on other diseases. In addition, this approach supports exploratory investigation through the review of meta paths that connect drugs with diseases. This knowledge-driven approach is an effective drug repurposing strategy supporting large-scale prediction and the investigation of case studies.
Collapse
Affiliation(s)
| | | | - Bo Jin
- Dalian University of Technology, China
| | | | | | | |
Collapse
|
28
|
SYBA: Bayesian estimation of synthetic accessibility of organic compounds. J Cheminform 2020; 12:35. [PMID: 33431015 PMCID: PMC7238540 DOI: 10.1186/s13321-020-00439-2] [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: 01/31/2020] [Accepted: 05/09/2020] [Indexed: 12/11/2022] Open
Abstract
SYBA (SYnthetic Bayesian Accessibility) is a fragment-based method for the rapid classification of organic compounds as easy- (ES) or hard-to-synthesize (HS). It is based on a Bernoulli naïve Bayes classifier that is used to assign SYBA score contributions to individual fragments based on their frequencies in the database of ES and HS molecules. SYBA was trained on ES molecules available in the ZINC15 database and on HS molecules generated by the Nonpher methodology. SYBA was compared with a random forest, that was utilized as a baseline method, as well as with other two methods for synthetic accessibility assessment: SAScore and SCScore. When used with their suggested thresholds, SYBA improves over random forest classification, albeit marginally, and outperforms SAScore and SCScore. However, upon the optimization of SAScore threshold (that changes from 6.0 to – 4.5), SAScore yields similar results as SYBA. Because SYBA is based merely on fragment contributions, it can be used for the analysis of the contribution of individual molecular parts to compound synthetic accessibility. SYBA is publicly available at https://github.com/lich-uct/syba under the GNU General Public License.
Collapse
|
29
|
Batool M, Ahmad B, Choi S. A Structure-Based Drug Discovery Paradigm. Int J Mol Sci 2019; 20:ijms20112783. [PMID: 31174387 PMCID: PMC6601033 DOI: 10.3390/ijms20112783] [Citation(s) in RCA: 298] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
Structure-based drug design is becoming an essential tool for faster and more cost-efficient lead discovery relative to the traditional method. Genomic, proteomic, and structural studies have provided hundreds of new targets and opportunities for future drug discovery. This situation poses a major problem: the necessity to handle the “big data” generated by combinatorial chemistry. Artificial intelligence (AI) and deep learning play a pivotal role in the analysis and systemization of larger data sets by statistical machine learning methods. Advanced AI-based sophisticated machine learning tools have a significant impact on the drug discovery process including medicinal chemistry. In this review, we focus on the currently available methods and algorithms for structure-based drug design including virtual screening and de novo drug design, with a special emphasis on AI- and deep-learning-based methods used for drug discovery.
Collapse
Affiliation(s)
- Maria Batool
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea.
| | - Bilal Ahmad
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea.
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea.
| |
Collapse
|
30
|
Schneider G, Clark DE. Automated De Novo Drug Design: Are We Nearly There Yet? Angew Chem Int Ed Engl 2019; 58:10792-10803. [PMID: 30730601 DOI: 10.1002/anie.201814681] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Indexed: 11/09/2022]
Abstract
Medicinal chemistry and, in particular, drug design have often been perceived as more of an art than a science. The many unknowns of human disease and the sheer complexity of chemical space render decision making in medicinal chemistry exceptionally demanding. Computational models can assist the medicinal chemist in this endeavour. Provided here is an overview of recent examples of automated de novo molecular design, a discussion of the concepts and computational approaches involved, and the daring prediction of some of the possibilities and limitations of drug design using machine intelligence.
Collapse
Affiliation(s)
- Gisbert Schneider
- ETH Zurich, Department of Chemistry and Applied Biosciences, RETHINK, Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - David E Clark
- Charles River, 6-9 Spire Green Centre, Harlow, Essex, CM19 5TR, UK
| |
Collapse
|
31
|
Schneider G, Clark DE. Automated De Novo Drug Design: Are We Nearly There Yet? Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201814681] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Gisbert Schneider
- ETH ZurichDepartment of Chemistry and Applied Biosciences, RETHINK Vladimir-Prelog-Weg 4 8093 Zurich Switzerland
| | - David E. Clark
- Charles River 6–9 Spire Green Centre Harlow Essex CM19 5TR UK
| |
Collapse
|
32
|
In silico structural elucidation of RNA-dependent RNA polymerase towards the identification of potential Crimean-Congo Hemorrhagic Fever Virus inhibitors. Sci Rep 2019; 9:6809. [PMID: 31048746 PMCID: PMC6497722 DOI: 10.1038/s41598-019-43129-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 04/17/2019] [Indexed: 01/05/2023] Open
Abstract
The Crimean-Congo Hemorrhagic Fever virus (CCHFV) is a segmented negative single-stranded RNA virus (-ssRNA) which causes severe hemorrhagic fever in humans with a mortality rate of ~50%. To date, no vaccine has been approved. Treatment is limited to supportive care with few investigational drugs in practice. Previous studies have identified viral RNA dependent RNA Polymerase (RdRp) as a potential drug target due to its significant role in viral replication and transcription. Since no crystal structure is available yet, we report the structural elucidation of CCHFV-RdRp by in-depth homology modeling. Even with low sequence identity, the generated model suggests a similar overall structure as previously reported RdRps. More specifically, the model suggests the presence of structural/functional conserved RdRp motifs for polymerase function, the configuration of uniform spatial arrangement of core RdRp sub-domains, and predicted positively charged entry/exit tunnels, as seen in sNSV polymerases. Extensive pharmacophore modeling based on per-residue energy contribution with investigational drugs allowed the concise mapping of pharmacophoric features and identified potential hits. The combination of pharmacophoric features with interaction energy analysis revealed functionally important residues in the conserved motifs together with in silico predicted common inhibitory binding modes with highly potent reference compounds.
Collapse
|
33
|
Abstract
De novo drug design aims to generate novel chemical compounds with desirable chemical and pharmacological properties from scratch using computer-based methods. Recently, deep generative neural networks have become a very active research frontier in de novo drug discovery, both in theoretical and in experimental evidence, shedding light on a promising new direction of automatic molecular generation and optimization. In this review, we discussed recent development of deep learning models for molecular generation and summarized them as four different generative architectures with four different optimization strategies. We also discussed future directions of deep generative models for de novo drug design.
Collapse
|
34
|
Athanasiou C, Cournia Z. From Computers to Bedside: Computational Chemistry Contributing to FDA Approval. BIOMOLECULAR SIMULATIONS IN STRUCTURE-BASED DRUG DISCOVERY 2018. [DOI: 10.1002/9783527806836.ch7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Christina Athanasiou
- Biomedical Research Foundation; Academy of Athens; 4 Soranou Ephessiou 11527 Athens Greece
| | - Zoe Cournia
- Biomedical Research Foundation; Academy of Athens; 4 Soranou Ephessiou 11527 Athens Greece
| |
Collapse
|
35
|
An approach towards enhancement of a screening library: The Next Generation Library Initiative (NGLI) at Bayer - against all odds? Drug Discov Today 2018; 24:668-672. [PMID: 30562586 DOI: 10.1016/j.drudis.2018.12.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/26/2018] [Accepted: 12/07/2018] [Indexed: 11/22/2022]
Abstract
Pharmaceutical companies often refer to 'screening their library' when performing high-throughput screening (HTS) on a corporate compound collection to identify lead structures for small-molecule drug discovery programs. Characteristics of such a library, including the size, chemical space covered, and physicochemical properties, often determine the success of a screening campaign. Therefore, strategies to maintain and enhance the overall quality of screening collections are crucial to stay competitive and to cope with the 'novelty erosion' that is observed gradually. The Next Generation Library Initiative (NGLI), the enhancement of Bayer's HTS collection by 500000 newly designed compounds within 5 years, is addressing exactly this challenge. Here, we describe this collaborative project, which involves all internal medicinal chemists in a crowd-sourcing approach, as well as selected external partners, to reach this ambitious goal.
Collapse
|
36
|
Merk D, Grisoni F, Friedrich L, Schneider G. Tuning artificial intelligence on the de novo design of natural-product-inspired retinoid X receptor modulators. Commun Chem 2018. [DOI: 10.1038/s42004-018-0068-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
37
|
Proschak E, Stark H, Merk D. Polypharmacology by Design: A Medicinal Chemist's Perspective on Multitargeting Compounds. J Med Chem 2018; 62:420-444. [PMID: 30035545 DOI: 10.1021/acs.jmedchem.8b00760] [Citation(s) in RCA: 293] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Multitargeting compounds comprising activity on more than a single biological target have gained remarkable relevance in drug discovery owing to the complexity of multifactorial diseases such as cancer, inflammation, or the metabolic syndrome. Polypharmacological drug profiles can produce additive or synergistic effects while reducing side effects and significantly contribute to the high therapeutic success of indispensable drugs such as aspirin. While their identification has long been the result of serendipity, medicinal chemistry now tends to design polypharmacology. Modern in vitro pharmacological methods and chemical probes allow a systematic search for rational target combinations and recent innovations in computational technologies, crystallography, or fragment-based design equip multitarget compound development with valuable tools. In this Perspective, we analyze the relevance of multiple ligands in drug discovery and the versatile toolbox to design polypharmacology. We conclude that despite some characteristic challenges remaining unresolved, designed polypharmacology holds enormous potential to secure future therapeutic innovation.
Collapse
Affiliation(s)
- Ewgenij Proschak
- Institute of Pharmaceutical Chemistry , Goethe University Frankfurt , Max-von-Laue-Strasse 9 , D-60438 Frankfurt , Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry , Heinrich Heine University Düsseldorf , Universitaetsstrasse 1 , D-40225 , Duesseldorf , Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry , Goethe University Frankfurt , Max-von-Laue-Strasse 9 , D-60438 Frankfurt , Germany.,Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences , Swiss Federal Institute of Technology (ETH) Zürich , Vladimir-Prelog-Weg 4 , CH-8093 Zürich , Switzerland
| |
Collapse
|
38
|
Hoffer L, Muller C, Roche P, Morelli X. Chemistry-driven Hit-to-lead Optimization Guided by Structure-based Approaches. Mol Inform 2018; 37:e1800059. [PMID: 30051601 DOI: 10.1002/minf.201800059] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 06/24/2018] [Indexed: 12/17/2022]
Abstract
For several decades, hit identification for drug discovery has been facilitated by developments in both fragment-based and high-throughput screening technologies. However, a major bottleneck in drug discovery projects continues to be the optimization of primary hits from screening campaigns in order to derive lead compounds. Computational chemistry or molecular modeling can play an important role during this hit-to-lead (H2L) stage by both suggesting putative optimizations and decreasing the number of compounds to be experimentally synthesized and evaluated. However, it is also crucial to consider the feasibility of organically synthesizing these virtually designed compounds. Furthermore, the generated molecules should have reasonable physicochemical properties and be medicinally relevant. This review focuses on chemistry-driven and structure-based computational methods that can be used to tackle the difficult problem of H2L optimization, with emphasis being placed on the strategy developed in our laboratory.
Collapse
Affiliation(s)
- Laurent Hoffer
- CNRS, Inserm, Institut Paoli-Calmettes, Aix-Marseille Univ, CRCM, Marseille, France
| | | | - Philippe Roche
- CNRS, Inserm, Institut Paoli-Calmettes, Aix-Marseille Univ, CRCM, Marseille, France
| | - Xavier Morelli
- CNRS, Inserm, Institut Paoli-Calmettes, Aix-Marseille Univ, CRCM, Marseille, France.,Institut Paoli-Calmettes, IPC Drug Discovery, Marseille, France
| |
Collapse
|
39
|
Artificial intelligence in drug design. SCIENCE CHINA-LIFE SCIENCES 2018; 61:1191-1204. [PMID: 30054833 DOI: 10.1007/s11427-018-9342-2] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/22/2018] [Indexed: 12/27/2022]
Abstract
Thanks to the fast improvement of the computing power and the rapid development of the computational chemistry and biology, the computer-aided drug design techniques have been successfully applied in almost every stage of the drug discovery and development pipeline to speed up the process of research and reduce the cost and risk related to preclinical and clinical trials. Owing to the development of machine learning theory and the accumulation of pharmacological data, the artificial intelligence (AI) technology, as a powerful data mining tool, has cut a figure in various fields of the drug design, such as virtual screening, activity scoring, quantitative structure-activity relationship (QSAR) analysis, de novo drug design, and in silico evaluation of absorption, distribution, metabolism, excretion and toxicity (ADME/T) properties. Although it is still challenging to provide a physical explanation of the AI-based models, it indeed has been acting as a great power to help manipulating the drug discovery through the versatile frameworks. Recently, due to the strong generalization ability and powerful feature extraction capability, deep learning methods have been employed in predicting the molecular properties as well as generating the desired molecules, which will further promote the application of AI technologies in the field of drug design.
Collapse
|
40
|
Hoffer L, Voitovich YV, Raux B, Carrasco K, Muller C, Fedorov AY, Derviaux C, Amouric A, Betzi S, Horvath D, Varnek A, Collette Y, Combes S, Roche P, Morelli X. Integrated Strategy for Lead Optimization Based on Fragment Growing: The Diversity-Oriented-Target-Focused-Synthesis Approach. J Med Chem 2018; 61:5719-5732. [PMID: 29883107 DOI: 10.1021/acs.jmedchem.8b00653] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Over the past few decades, hit identification has been greatly facilitated by advances in high-throughput and fragment-based screenings. One major hurdle remaining in drug discovery is process automation of hit-to-lead (H2L) optimization. Here, we report a time- and cost-efficient integrated strategy for H2L optimization as well as a partially automated design of potent chemical probes consisting of a focused-chemical-library design and virtual screening coupled with robotic diversity-oriented de novo synthesis and automated in vitro evaluation. The virtual library is generated by combining an activated fragment, corresponding to the substructure binding to the target, with a collection of functionalized building blocks using in silico encoded chemical reactions carefully chosen from a list of one-step organic transformations relevant in medicinal chemistry. The proof of concept was demonstrated using the optimization of bromodomain inhibitors as a test case, leading to the validation of several compounds with improved affinity by several orders of magnitude.
Collapse
Affiliation(s)
- Laurent Hoffer
- CRCM, CNRS, Inserm, Institut Paoli-Calmettes , Aix-Marseille University , 13009 Marseille , France
| | - Yuliia V Voitovich
- CRCM, CNRS, Inserm, Institut Paoli-Calmettes , Aix-Marseille University , 13009 Marseille , France.,Department of Organic Chemistry , Lobachevsky State University of Nizhni Novgorod , 23 Gagarin Avenue , 603950 Nizhni Novgorod , Russia
| | - Brigitt Raux
- CRCM, CNRS, Inserm, Institut Paoli-Calmettes , Aix-Marseille University , 13009 Marseille , France
| | - Kendall Carrasco
- CRCM, CNRS, Inserm, Institut Paoli-Calmettes , Aix-Marseille University , 13009 Marseille , France
| | - Christophe Muller
- IPC Drug Discovery Platform , Institut Paoli-Calmettes , 232 Boulevard de Sainte-Marguerite , 13009 Marseille , France
| | - Aleksey Y Fedorov
- Department of Organic Chemistry , Lobachevsky State University of Nizhni Novgorod , 23 Gagarin Avenue , 603950 Nizhni Novgorod , Russia
| | - Carine Derviaux
- IPC Drug Discovery Platform , Institut Paoli-Calmettes , 232 Boulevard de Sainte-Marguerite , 13009 Marseille , France
| | - Agnès Amouric
- CRCM, CNRS, Inserm, Institut Paoli-Calmettes , Aix-Marseille University , 13009 Marseille , France.,IPC Drug Discovery Platform , Institut Paoli-Calmettes , 232 Boulevard de Sainte-Marguerite , 13009 Marseille , France
| | - Stéphane Betzi
- CRCM, CNRS, Inserm, Institut Paoli-Calmettes , Aix-Marseille University , 13009 Marseille , France
| | - Dragos Horvath
- Laboratoire de Chemoinformatique, CNRS UMR7140 , 1 rue Blaise Pascal , 67000 Strasbourg , France
| | - Alexandre Varnek
- Laboratoire de Chemoinformatique, CNRS UMR7140 , 1 rue Blaise Pascal , 67000 Strasbourg , France
| | - Yves Collette
- CRCM, CNRS, Inserm, Institut Paoli-Calmettes , Aix-Marseille University , 13009 Marseille , France.,IPC Drug Discovery Platform , Institut Paoli-Calmettes , 232 Boulevard de Sainte-Marguerite , 13009 Marseille , France
| | - Sébastien Combes
- CRCM, CNRS, Inserm, Institut Paoli-Calmettes , Aix-Marseille University , 13009 Marseille , France
| | - Philippe Roche
- CRCM, CNRS, Inserm, Institut Paoli-Calmettes , Aix-Marseille University , 13009 Marseille , France
| | - Xavier Morelli
- CRCM, CNRS, Inserm, Institut Paoli-Calmettes , Aix-Marseille University , 13009 Marseille , France.,IPC Drug Discovery Platform , Institut Paoli-Calmettes , 232 Boulevard de Sainte-Marguerite , 13009 Marseille , France
| |
Collapse
|
41
|
Merk D, Grisoni F, Friedrich L, Gelzinyte E, Schneider G. Computer-Assisted Discovery of Retinoid X Receptor Modulating Natural Products and Isofunctional Mimetics. J Med Chem 2018; 61:5442-5447. [DOI: 10.1021/acs.jmedchem.8b00494] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Daniel Merk
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Francesca Grisoni
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza, 1, IT-20126 Milan, Italy
| | - Lukas Friedrich
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Elena Gelzinyte
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Gisbert Schneider
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| |
Collapse
|
42
|
Schneider P, Schneider G. Polypharmacological Drug−target Inference for Chemogenomics. Mol Inform 2018; 37:e1800050. [DOI: 10.1002/minf.201800050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 04/24/2018] [Indexed: 01/10/2023]
Affiliation(s)
- Petra Schneider
- Department of Chemistry and Applied Biosciences; Swiss Federal Institute of Technology (ETH); Vladimir-Prelog-Weg 4 CH-8093 Zurich Switzerland
| | - Gisbert Schneider
- Department of Chemistry and Applied Biosciences; Swiss Federal Institute of Technology (ETH); Vladimir-Prelog-Weg 4 CH-8093 Zurich Switzerland
| |
Collapse
|
43
|
Hachmann J, Afzal MAF, Haghighatlari M, Pal Y. Building and deploying a cyberinfrastructure for the data-driven design of chemical systems and the exploration of chemical space. MOLECULAR SIMULATION 2018. [DOI: 10.1080/08927022.2018.1471692] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Johannes Hachmann
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York , Buffalo, NY, USA
- Computational and Data-Enabled Science and Engineering Graduate Program, University at Buffalo, The State University of New York , Buffalo, NY, USA
- New York State Center of Excellence in Materials Informatics , Buffalo, NY, USA
| | - Mohammad Atif Faiz Afzal
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York , Buffalo, NY, USA
| | - Mojtaba Haghighatlari
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York , Buffalo, NY, USA
| | - Yudhajit Pal
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York , Buffalo, NY, USA
| |
Collapse
|
44
|
Abstract
The term drug design describes the search of novel compounds with biological activity, on a systematic basis. In its most common form, it involves modification of a known active scaffold or linking known active scaffolds, although de novo drug design (i.e., from scratch) is also possible. Though highly interrelated, identification of active scaffolds should be conceptually separated from drug design. Traditionally, the drug design process has focused on the molecular determinants of the interactions between the drug and its known or intended molecular target. Nevertheless, current drug design also takes into consideration other relevant processes than influence drug efficacy and safety (e.g., bioavailability, metabolic stability, interaction with antitargets).This chapter provides an overview on possible approaches to identify active scaffolds (including in silico approximations to approach that task) and computational methods to guide the subsequent optimization process. It also discusses in which situations each of the overviewed techniques is more appropriate.
Collapse
Affiliation(s)
- Alan Talevi
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Faculty of Exact Sciences, National University of La Plata (UNLP), Buenos Aires, Argentina.
- Argentinean National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
45
|
Merk D, Friedrich L, Grisoni F, Schneider G. De Novo Design of Bioactive Small Molecules by Artificial Intelligence. Mol Inform 2018; 37:1700153. [PMID: 29319225 PMCID: PMC5838524 DOI: 10.1002/minf.201700153] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 12/20/2017] [Indexed: 11/12/2022]
Abstract
Generative artificial intelligence offers a fresh view on molecular design. We present the first-time prospective application of a deep learning model for designing new druglike compounds with desired activities. For this purpose, we trained a recurrent neural network to capture the constitution of a large set of known bioactive compounds represented as SMILES strings. By transfer learning, this general model was fine-tuned on recognizing retinoid X and peroxisome proliferator-activated receptor agonists. We synthesized five top-ranking compounds designed by the generative model. Four of the compounds revealed nanomolar to low-micromolar receptor modulatory activity in cell-based assays. Apparently, the computational model intrinsically captured relevant chemical and biological knowledge without the need for explicit rules. The results of this study advocate generative artificial intelligence for prospective de novo molecular design, and demonstrate the potential of these methods for future medicinal chemistry.
Collapse
Affiliation(s)
- Daniel Merk
- Department of Chemistry and Applied BiosciencesSwiss Federal Institute of Technology (ETH)Vladimir-Prelog-Weg 4, CH-8093ZurichSwitzerland
| | - Lukas Friedrich
- Department of Chemistry and Applied BiosciencesSwiss Federal Institute of Technology (ETH)Vladimir-Prelog-Weg 4, CH-8093ZurichSwitzerland
| | - Francesca Grisoni
- Department of Chemistry and Applied BiosciencesSwiss Federal Institute of Technology (ETH)Vladimir-Prelog-Weg 4, CH-8093ZurichSwitzerland
- Department of Earth and Environmental SciencesUniversity of Milano-BicoccaP.za della Scienza, 1, IT-20126MilanItaly
| | - Gisbert Schneider
- Department of Chemistry and Applied BiosciencesSwiss Federal Institute of Technology (ETH)Vladimir-Prelog-Weg 4, CH-8093ZurichSwitzerland
| |
Collapse
|
46
|
Abstract
In the history of therapeutics, covalent drugs occupy a very distinct category. While representing a significant fraction of the drugs on the market, very few have been deliberately designed to interact covalently with their biological target. In this review, the prevalence of covalent drugs will first be briefly covered, followed by an introduction to their mechanisms of action and more detailed discussions of their discovery and the development of safe and efficient covalent enzyme inhibitors. All stages of a drug discovery program will be covered, from target considerations to lead optimization, strategies to tune reactivity and computational methods. The goal of this article is to provide an overview of the field and to outline good practices that are needed for the proper assessment and development of covalent inhibitors as well as a good understanding of the potential and limitations of current computational methods for the design of covalent drugs.
Collapse
Affiliation(s)
- Stephane De Cesco
- Department of Chemistry, McGill University, 801 Sherbrooke St. W., Montréal, Québec H3A 0B8, Canada
| | - Jerry Kurian
- Department of Chemistry, McGill University, 801 Sherbrooke St. W., Montréal, Québec H3A 0B8, Canada
| | - Caroline Dufresne
- Department of Chemistry, McGill University, 801 Sherbrooke St. W., Montréal, Québec H3A 0B8, Canada
| | - Anthony K Mittermaier
- Department of Chemistry, McGill University, 801 Sherbrooke St. W., Montréal, Québec H3A 0B8, Canada
| | - Nicolas Moitessier
- Department of Chemistry, McGill University, 801 Sherbrooke St. W., Montréal, Québec H3A 0B8, Canada.
| |
Collapse
|
47
|
Pottel J, Moitessier N. Customizable Generation of Synthetically Accessible, Local Chemical Subspaces. J Chem Inf Model 2017; 57:454-467. [DOI: 10.1021/acs.jcim.6b00648] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Joshua Pottel
- Department of Chemistry, McGill University, 801
Sherbrooke Street W., Montréal, Québec, Canada H3A 0B8
| | - Nicolas Moitessier
- Department of Chemistry, McGill University, 801
Sherbrooke Street W., Montréal, Québec, Canada H3A 0B8
| |
Collapse
|
48
|
|
49
|
Friedrich L, Rodrigues T, Neuhaus CS, Schneider P, Schneider G. From Complex Natural Products to Simple Synthetic Mimetics by Computational De Novo Design. Angew Chem Int Ed Engl 2016; 55:6789-92. [DOI: 10.1002/anie.201601941] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/03/2016] [Indexed: 11/08/2022]
Affiliation(s)
- Lukas Friedrich
- Department of Chemistry and Applied Biosciences; Swiss Federal Institute of Technology (ETH); Vladimir-Prelog-Weg 4 8093 Zurich Switzerland
| | - Tiago Rodrigues
- Department of Chemistry and Applied Biosciences; Swiss Federal Institute of Technology (ETH); Vladimir-Prelog-Weg 4 8093 Zurich Switzerland
| | - Claudia S. Neuhaus
- Department of Chemistry and Applied Biosciences; Swiss Federal Institute of Technology (ETH); Vladimir-Prelog-Weg 4 8093 Zurich Switzerland
| | - Petra Schneider
- Department of Chemistry and Applied Biosciences; Swiss Federal Institute of Technology (ETH); Vladimir-Prelog-Weg 4 8093 Zurich Switzerland
- inSili.com GmbH; Segantinisteig 3 8049 Zurich Switzerland
| | - Gisbert Schneider
- Department of Chemistry and Applied Biosciences; Swiss Federal Institute of Technology (ETH); Vladimir-Prelog-Weg 4 8093 Zurich Switzerland
| |
Collapse
|
50
|
Friedrich L, Rodrigues T, Neuhaus CS, Schneider P, Schneider G. Von komplexen Naturstoffen zu synthetisch leicht zugänglichen Mimetika mithilfe von computergestütztem De-novo-Design. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201601941] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Lukas Friedrich
- Department für Chemie und Angewandte Biowissenschaften; Eidgenössische Technische Hochschule; Vladimir-Prelog-Weg 4 8093 Zürich Schweiz
| | - Tiago Rodrigues
- Department für Chemie und Angewandte Biowissenschaften; Eidgenössische Technische Hochschule; Vladimir-Prelog-Weg 4 8093 Zürich Schweiz
| | - Claudia S. Neuhaus
- Department für Chemie und Angewandte Biowissenschaften; Eidgenössische Technische Hochschule; Vladimir-Prelog-Weg 4 8093 Zürich Schweiz
| | - Petra Schneider
- Department für Chemie und Angewandte Biowissenschaften; Eidgenössische Technische Hochschule; Vladimir-Prelog-Weg 4 8093 Zürich Schweiz
- inSili.com GmbH; Segantinisteig 3 8049 Zürich Schweiz
| | - Gisbert Schneider
- Department für Chemie und Angewandte Biowissenschaften; Eidgenössische Technische Hochschule; Vladimir-Prelog-Weg 4 8093 Zürich Schweiz
| |
Collapse
|