1
|
Xing F, Su HY, Zhong HY, Li YZ, Zhang YY, Chen L, Zhou XL. Synthesis and biological evaluation of lappaconitine analogues as potential anti-neuroinflammatory agents by side chain modification and scaffold hopping strategy. Bioorg Med Chem 2025; 117:118012. [PMID: 39608210 DOI: 10.1016/j.bmc.2024.118012] [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: 08/16/2024] [Revised: 11/10/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024]
Abstract
Neuroinflammation mediated by microglia is widely recognized as a key pathophysiological mechanism in neurodegenerative diseases. Lappaconitine (LA) is a natural C18-diterpenoid alkaloid isolated from Aconitum sinomontanum Nakai, and previous study showed that LA and its derivatives inhibited lipopolysaccharide (LPS)-induced nitric oxide (NO) production in RAW264.7 cells. However, the anti-neuroinflammatory effects of LA and its derivatives on microglia are still not clear. Here, LA analogues were designed and synthesized, and the anti-neuroinflammatory activity of the synthesized compounds was screened using LPS-induced overexpression of NO in BV-2 microglia. The screening results showed that compound 10 displayed the highest ability to inhibit NO production (IC50 = 9.98 ± 1.6 µM). Mechanistic investigations revealed that compound 10 attenuated LPS-activated neuroinflammation through suppression of TLR4/MyD88/NF-κB pathway in BV-2 microglia. Acute toxicity assays showed that compound 10 (LD50 = 508.1 mg/kg) was safer relative to LA (LD50 = 30.6 mg/kg). Collectively, our findings show that compound 10 could have potential as anti-neuroinflammatory agents.
Collapse
Affiliation(s)
- Feng Xing
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Hong-Yi Su
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - He-Yang Zhong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yu-Zhu Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yin-Yong Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lin Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Xian-Li Zhou
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China; School of Life Science and Engineering, Yibin Institute of Southwest Jiaotong University, Yibin 644000, China.
| |
Collapse
|
2
|
Dai J, Zhang Y, Shi C, Liu Y, Xiu P, Wang Y. BEGAN: Boltzmann-Reweighted Data Augmentation for Enhanced GAN-Based Molecule Design in Insect Pheromone Receptors. J Phys Chem B 2024; 128:11666-11675. [PMID: 39540850 DOI: 10.1021/acs.jpcb.4c06729] [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: 11/16/2024]
Abstract
Identifying small molecules that bind strongly to target proteins in rational molecular design is crucial. Machine learning techniques, such as generative adversarial networks (GAN), are now essential tools for generating such molecules. In this study, we present an enhanced method for molecule generation using objective-reinforced GANs. Specifically, we introduce BEGAN (Boltzmann-enhanced GAN), a novel approach that adjusts molecule occurrence frequencies during training based on the Boltzmann distribution exp(-ΔU/τ), where ΔU represents the estimated binding free energy derived from docking algorithms and τ is a temperature-related scaling hyperparameter. This Boltzmann reweighting process shifts the generation process toward molecules with higher binding affinities, allowing the GAN to explore molecular spaces with superior binding properties. The reweighting process can also be refined through multiple iterations without altering the overall distribution shape. To validate our approach, we apply it to the design of sex pheromone analogs targeting Spodoptera frugiperda pheromone receptor SfruOR16, illustrating that the Boltzmann reweighting significantly increases the likelihood of generating promising sex pheromone analogs with improved binding affinities to SfruOR16, further supported by atomistic molecular dynamics simulations. Furthermore, we conduct a comprehensive investigation into parameter dependencies and propose a reasonable range for the hyperparameter τ. Our method offers a promising approach for optimizing molecular generation for enhanced protein binding, potentially increasing the efficiency of molecular discovery pipelines.
Collapse
Affiliation(s)
- Jialei Dai
- College of Life Sciences, Zhejiang University, Hangzhou 310027, China
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China
| | - Yutong Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Chen Shi
- College of Life Sciences, Zhejiang University, Hangzhou 310027, China
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China
| | - Yang Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Peng Xiu
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China
| | - Yong Wang
- College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
3
|
Zhang O, Lin H, Zhang H, Zhao H, Huang Y, Hsieh CY, Pan P, Hou T. Deep Lead Optimization: Leveraging Generative AI for Structural Modification. J Am Chem Soc 2024; 146:31357-31370. [PMID: 39499822 DOI: 10.1021/jacs.4c11686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
The integration of deep learning-based molecular generation models into drug discovery has garnered significant attention for its potential to expedite the development process. Central to this is lead optimization, a critical phase where existing molecules are refined into viable drug candidates. As various methods for deep lead optimization continue to emerge, it is essential to classify these approaches more clearly. We categorize lead optimization methods into two main types: goal-directed and structure-directed. Our focus is on structure-directed optimization, which, while highly relevant to practical applications, is less explored compared to goal-directed methods. Through a systematic review of conventional computational approaches, we identify four tasks specific to structure-directed optimization: fragment replacement, linker design, scaffold hopping, and side-chain decoration. We discuss the motivations, training data construction, and current developments for each of these tasks. Additionally, we use classical optimization taxonomy to classify both goal-directed and structure-directed methods, highlighting their challenges and future development prospects. Finally, we propose a reference protocol for experimental chemists to effectively utilize Generative AI (GenAI)-based tools in structural modification tasks, bridging the gap between methodological advancements and practical applications.
Collapse
Affiliation(s)
- Odin Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Haitao Lin
- AI Lab, Research Center for Industries of the Future, Westlake University, Hangzhou 310024, Zhejiang, China
| | - Hui Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Huifeng Zhao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Yufei Huang
- AI Lab, Research Center for Industries of the Future, Westlake University, Hangzhou 310024, Zhejiang, China
| | - Chang-Yu Hsieh
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Peichen Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| |
Collapse
|
4
|
Flores-Hernandez H, Martinez-Ledesma E. A systematic review of deep learning chemical language models in recent era. J Cheminform 2024; 16:129. [PMID: 39558376 PMCID: PMC11571686 DOI: 10.1186/s13321-024-00916-y] [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: 07/26/2024] [Accepted: 10/17/2024] [Indexed: 11/20/2024] Open
Abstract
Discovering new chemical compounds with specific properties can provide advantages for fields that rely on materials for their development, although this task comes at a high cost in terms of complexity and resources. Since the beginning of the data age, deep learning techniques have revolutionized the process of designing molecules by analyzing and learning from representations of molecular data, greatly reducing the resources and time involved. Various deep learning approaches have been developed to date, using a variety of architectures and strategies, in order to explore the extensive and discontinuous chemical space, providing benefits for generating compounds with specific properties. In this study, we present a systematic review that offers a statistical description and comparison of the strategies utilized to generate molecules through deep learning techniques, utilizing the metrics proposed in Molecular Sets (MOSES) or Guacamol. The study included 48 articles retrieved from a query-based search of Scopus and Web of Science and 25 articles retrieved from citation search, yielding a total of 72 retrieved articles, of which 62 correspond to chemical language models approaches to molecule generation and other 10 retrieved articles correspond to molecular graph representations. Transformers, recurrent neural networks (RNNs), generative adversarial networks (GANs), Structured Space State Sequence (S4) models, and variational autoencoders (VAEs) are considered the main deep learning architectures used for molecule generation in the set of retrieved articles. In addition, transfer learning, reinforcement learning, and conditional learning are the most employed techniques for biased model generation and exploration of specific chemical space regions. Finally, this analysis focuses on the central themes of molecular representation, databases, training dataset size, validity-novelty trade-off, and performance of unbiased and biased chemical language models. These themes were selected to conduct a statistical analysis utilizing graphical representation and statistical tests. The resulting analysis reveals the main challenges, advantages, and opportunities in the field of chemical language models over the past four years.
Collapse
Affiliation(s)
- Hector Flores-Hernandez
- Tecnológico de Monterrey, School of Engineering and Sciences, Monterrey, 64710, Nuevo León, México
| | - Emmanuel Martinez-Ledesma
- Tecnológico de Monterrey, School of Medicine and Health Sciences, Monterrey, 64710, Nuevo León, México.
- Institute for Obesity Research, Tecnológico de Monterrey, Monterrey, 64710, Nuevo León, México.
| |
Collapse
|
5
|
Mishra A, Thakur A, Sharma R, Onuku R, Kaur C, Liou JP, Hsu SP, Nepali K. Scaffold hopping approaches for dual-target antitumor drug discovery: opportunities and challenges. Expert Opin Drug Discov 2024; 19:1355-1381. [PMID: 39420580 DOI: 10.1080/17460441.2024.2409674] [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: 06/07/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Scaffold hopping has emerged as a practical tactic to enrich the synthetic bank of small molecule antitumor agents. Specifically, it enables the chemist to refine the lead compound's pharmacodynamic, pharmacokinetic, and physiochemical properties. Scaffold hopping opens up fresh molecular territory beyond established patented chemical domains. AREA COVERED The authors present the scaffold hopping-based drug design strategies for dual inhibitory antitumor structural templates in this review. Minor modifications, structure rigidification and simplification (ring-closing and opening), and complete structural overhauls were the strategies employed by the medicinal chemist to generate a library of bifunctional inhibitors. In addition, the review presents an overview of the computational methods of scaffold hopping (software and programs) and organopalladium catalysis leveraged for the synthesis of templates designed via scaffold hopping. EXPERT OPINION The medicinal chemist has demonstrated remarkable prowess in furnishing dual inhibitory antitumor chemical architectures. Scaffold hopping-based drug design strategies have yielded a plethora of pharmacodynamically superior dual modulatory antitumor agents. An integrated approach involving computational advancements, synthetic methodology advancements, and conventional drug design strategies is required to increase the number of scaffold-hopping-assisted drug discovery campaigns.
Collapse
Affiliation(s)
- Anshul Mishra
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Raphael Onuku
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Charanjit Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Sung-Po Hsu
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| |
Collapse
|
6
|
Chou CL, Lin CT, Kao CT, Lin CC. A Novel Rational PROTACs Design and Validation via AI-Driven Drug Design Approach. ACS OMEGA 2024; 9:38371-38384. [PMID: 39310161 PMCID: PMC11411543 DOI: 10.1021/acsomega.3c10183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 07/31/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024]
Abstract
The rational design of novel drug candidates presents a formidable challenge in modern drug discovery. Proteolysis-targeting chimeras (PROTACs) drug design is particularly demanding due to their limited crystal structure availability and design of a viable small molecule to bridge the protein of interest (POI) and ubiquitin-protein ligase (E3). An integrated approach that combines superimposition techniques and deep neural networks is demonstrated in this study to leverage the power of deep learning and structural biology to generate structurally diverse molecules with enhanced binding affinities. The superimposition technique ensures the congruence of initial and new protein-ligand pairs, which are evaluated via subsequent comprehensive screening using the root-mean-square deviation (RMSD), binding free energy (BFE), and buried solvent-accessible surface area (SASA). The final candidates are subjected to the incorporation of molecular dynamics (MD) and free energy perturbation (FEP) simulations to provide a quantitative evaluation of relative binding energies, reinforcing the efficacy and reliability of the generated molecules. The outcomes of the generated novel PROTACs molecules exhibit comparable structural attributes while demonstrating superior binding affinities within the binding pockets when contrasted with those of the established cocrystal ternary complexes. To enhance the generalizability of the workflow, we chose the ternary structure of the cellular inhibitor of apoptosis protein 1 (cIAP1) and Bruton's Tyrosine Kinase (BTK) for validating the chemical properties generated from the processes. The new linker molecules additionally showed superior affinity from the simulations. In summary, this methodology serves as an effective workflow to align computational predictions with current limitations, thereby introducing a novel paradigm in AI-driven drug design.
Collapse
Affiliation(s)
| | - Chieh-Te Lin
- Department
of Biomedical Engineering, University of
California Davis, Davis, California 95616, United States
| | | | | |
Collapse
|
7
|
He J, Tibo A, Janet JP, Nittinger E, Tyrchan C, Czechtizky W, Engkvist O. Evaluation of reinforcement learning in transformer-based molecular design. J Cheminform 2024; 16:95. [PMID: 39118113 PMCID: PMC11312936 DOI: 10.1186/s13321-024-00887-0] [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: 03/15/2024] [Accepted: 07/21/2024] [Indexed: 08/10/2024] Open
Abstract
Designing compounds with a range of desirable properties is a fundamental challenge in drug discovery. In pre-clinical early drug discovery, novel compounds are often designed based on an already existing promising starting compound through structural modifications for further property optimization. Recently, transformer-based deep learning models have been explored for the task of molecular optimization by training on pairs of similar molecules. This provides a starting point for generating similar molecules to a given input molecule, but has limited flexibility regarding user-defined property profiles. Here, we evaluate the effect of reinforcement learning on transformer-based molecular generative models. The generative model can be considered as a pre-trained model with knowledge of the chemical space close to an input compound, while reinforcement learning can be viewed as a tuning phase, steering the model towards chemical space with user-specific desirable properties. The evaluation of two distinct tasks-molecular optimization and scaffold discovery-suggest that reinforcement learning could guide the transformer-based generative model towards the generation of more compounds of interest. Additionally, the impact of pre-trained models, learning steps and learning rates are investigated.Scientific contributionOur study investigates the effect of reinforcement learning on a transformer-based generative model initially trained for generating molecules similar to starting molecules. The reinforcement learning framework is applied to facilitate multiparameter optimisation of starting molecules. This approach allows for more flexibility for optimizing user-specific property profiles and helps finding more ideas of interest.
Collapse
Affiliation(s)
- Jiazhen He
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| | - Alessandro Tibo
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Jon Paul Janet
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Eva Nittinger
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Christian Tyrchan
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Werngard Czechtizky
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ola Engkvist
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
- Department of Computer Science and Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
8
|
Ahmad S, Raza K. An extensive review on lung cancer therapeutics using machine learning techniques: state-of-the-art and perspectives. J Drug Target 2024; 32:635-646. [PMID: 38662768 DOI: 10.1080/1061186x.2024.2347358] [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: 02/10/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
There are over 100 types of human cancer, accounting for millions of deaths every year. Lung cancer alone claims over 1.8 million lives per year and is expected to surpass 3.2 million by 2050, which underscores the urgent need for rapid drug development and repurposing initiatives. The application of AI emerges as a pivotal solution to developing anti-cancer therapeutics. This state-of-the-art review aims to explore the various applications of AI in lung cancer therapeutics. Predictive models can analyse large datasets, including clinical data, genetic information, and treatment outcomes, for novel drug design and to generate personalised treatment recommendations, potentially optimising therapeutic strategies, enhancing treatment efficacy, and minimising adverse effects. A thorough literature review study was conducted based on articles indexed in PubMed and Scopus. We compiled the use of various machine learning approaches, including CNN, RNN, GAN, VAEs, and other AI techniques, enhancing efficiency with accuracy exceeding 95%, which is validated through a computer-aided drug design process. AI can revolutionise lung cancer therapeutics, streamlining processes and saving biological scientists' time and effort-however, further research is needed to overcome challenges and fully unlock AI's potential in Lung Cancer Therapeutics.
Collapse
Affiliation(s)
- Shaban Ahmad
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Khalid Raza
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
9
|
Luong KD, Singh A. Application of Transformers in Cheminformatics. J Chem Inf Model 2024; 64:4392-4409. [PMID: 38815246 PMCID: PMC11167597 DOI: 10.1021/acs.jcim.3c02070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/05/2024] [Accepted: 05/06/2024] [Indexed: 06/01/2024]
Abstract
By accelerating time-consuming processes with high efficiency, computing has become an essential part of many modern chemical pipelines. Machine learning is a class of computing methods that can discover patterns within chemical data and utilize this knowledge for a wide variety of downstream tasks, such as property prediction or substance generation. The complex and diverse chemical space requires complex machine learning architectures with great learning power. Recently, learning models based on transformer architectures have revolutionized multiple domains of machine learning, including natural language processing and computer vision. Naturally, there have been ongoing endeavors in adopting these techniques to the chemical domain, resulting in a surge of publications within a short period. The diversity of chemical structures, use cases, and learning models necessitate a comprehensive summarization of existing works. In this paper, we review recent innovations in adapting transformers to solve learning problems in chemistry. Because chemical data is diverse and complex, we structure our discussion based on chemical representations. Specifically, we highlight the strengths and weaknesses of each representation, the current progress of adapting transformer architectures, and future directions.
Collapse
Affiliation(s)
- Kha-Dinh Luong
- Department of Computer Science, University of California Santa Barbara, Santa Barbara, CA 93106, United States
| | - Ambuj Singh
- Department of Computer Science, University of California Santa Barbara, Santa Barbara, CA 93106, United States
| |
Collapse
|
10
|
Qian Y, Shi M, Zhang Q. CONSMI: Contrastive Learning in the Simplified Molecular Input Line Entry System Helps Generate Better Molecules. Molecules 2024; 29:495. [PMID: 38276573 PMCID: PMC10821140 DOI: 10.3390/molecules29020495] [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: 11/15/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
In recent years, the application of deep learning in molecular de novo design has gained significant attention. One successful approach involves using SMILES representations of molecules and treating the generation task as a text generation problem, yielding promising results. However, the generation of more effective and novel molecules remains a key research area. Due to the fact that a molecule can have multiple SMILES representations, it is not sufficient to consider only one of them for molecular generation. To make up for this deficiency, and also motivated by the advancements in contrastive learning in natural language processing, we propose a contrastive learning framework called CONSMI to learn more comprehensive SMILES representations. This framework leverages different SMILES representations of the same molecule as positive examples and other SMILES representations as negative examples for contrastive learning. The experimental results of generation tasks demonstrate that CONSMI significantly enhances the novelty of generated molecules while maintaining a high validity. Moreover, the generated molecules have similar chemical properties compared to the original dataset. Additionally, we find that CONSMI can achieve favorable results in classifier tasks, such as the compound-protein interaction task.
Collapse
Affiliation(s)
| | | | - Qian Zhang
- School of Computer Science and Technology, Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, East China Normal University, 3663 North Zhongshan Road, Putuo District, Shanghai 200062, China; (Y.Q.); (M.S.)
| |
Collapse
|
11
|
Xu M, Chen H. Tree-Invent: A Novel Multipurpose Molecular Generative Model Constrained with a Topological Tree. J Chem Inf Model 2023; 63:7067-7082. [PMID: 37962855 DOI: 10.1021/acs.jcim.3c01626] [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: 11/15/2023]
Abstract
De novo molecular design plays an important role in drug discovery. Here, a novel generative model, Tree-Invent, was proposed to integrate topological constraints in the generation of a molecular graph. In this model, a molecular graph is represented as a topological tree in which a ring system, a nonring atom, and a chemical bond are regarded as the ring node, single node, and edge, respectively. The molecule generation is driven by three independent submodels for carrying out operations of node addition, ring generation, and node connection. One unique feature of the generative model is that the topological tree structure can be specified as a constraint for structure generation, which provides more precise control of structure generation. Combined with reinforcement learning, the Tree-Invent model could efficiently explore targeted chemical space. Moreover, the Tree-Invent model is flexible enough to be used in versatile molecule design settings such as scaffold decoration, scaffold hopping, and linker generation.
Collapse
Affiliation(s)
- Mingyuan Xu
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, Guangdong 510005, China
| | - Hongming Chen
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, Guangdong 510005, China
| |
Collapse
|
12
|
Erikawa D, Yasuo N, Suzuki T, Nakamura S, Sekijima M. Gargoyles: An Open Source Graph-Based Molecular Optimization Method Based on Deep Reinforcement Learning. ACS OMEGA 2023; 8:37431-37441. [PMID: 37841174 PMCID: PMC10568706 DOI: 10.1021/acsomega.3c05430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023]
Abstract
Automatic optimization methods for compounds in the vast compound space are important for drug discovery and material design. Several machine learning-based molecular generative models for drug discovery have been proposed, but most of these methods generate compounds from scratch and are not suitable for exploring and optimizing user-defined compounds. In this study, we developed a compound optimization method based on molecular graphs using deep reinforcement learning. This method searches for compounds on a fragment-by-fragment basis and at high density by generating fragments to be added atom by atom. Experimental results confirmed that the quantum electrodynamics (QED), the optimization target set in this study, was enhanced by searching around the starting compound. As a use case, we successfully enhanced the activity of a compound by targeting dopamine receptor D2 (DRD2). This means that the generated compounds are not structurally dissimilar from the starting compounds, as well as increasing their activity, indicating that this method is suitable for optimizing molecules from a given compound. The source code is available at https://github.com/sekijima-lab/GARGOYLES.
Collapse
Affiliation(s)
- Daiki Erikawa
- Department
of Computer Science, Tokyo Institute of
Technology, 4259-J3-23, Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Nobuaki Yasuo
- Academy
for Convergence of Materials and Informatics (TAC-MI), Tokyo Institute of Technology, S6-23, Ookayama, Meguro-ku, Tokyo 152-8550, Japan
| | - Takamasa Suzuki
- Department
of Computer Science, Tokyo Institute of
Technology, 4259-J3-23, Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Shogo Nakamura
- Department
of Life Science and Technology, Tokyo Institute
of Technology, 4259-J3-23, Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Masakazu Sekijima
- Department
of Computer Science, Tokyo Institute of
Technology, 4259-J3-23, Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| |
Collapse
|
13
|
Hu C, Li S, Yang C, Chen J, Xiong Y, Fan G, Liu H, Hong L. ScaffoldGVAE: scaffold generation and hopping of drug molecules via a variational autoencoder based on multi-view graph neural networks. J Cheminform 2023; 15:91. [PMID: 37794460 PMCID: PMC10548653 DOI: 10.1186/s13321-023-00766-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023] Open
Abstract
In recent years, drug design has been revolutionized by the application of deep learning techniques, and molecule generation is a crucial aspect of this transformation. However, most of the current deep learning approaches do not explicitly consider and apply scaffold hopping strategy when performing molecular generation. In this work, we propose ScaffoldGVAE, a variational autoencoder based on multi-view graph neural networks, for scaffold generation and scaffold hopping of drug molecules. The model integrates several important components, such as node-central and edge-central message passing, side-chain embedding, and Gaussian mixture distribution of scaffolds. To assess the efficacy of our model, we conduct a comprehensive evaluation and comparison with baseline models based on seven general generative model evaluation metrics and four scaffold hopping generative model evaluation metrics. The results demonstrate that ScaffoldGVAE can explore the unseen chemical space and generate novel molecules distinct from known compounds. Especially, the scaffold hopped molecules generated by our model are validated by the evaluation of GraphDTA, LeDock, and MM/GBSA. The case study of generating inhibitors of LRRK2 for the treatment of PD further demonstrates the effectiveness of ScaffoldGVAE in generating novel compounds through scaffold hopping. This novel approach can also be applied to other protein targets of various diseases, thereby contributing to the future development of new drugs. Source codes and data are available at https://github.com/ecust-hc/ScaffoldGVAE .
Collapse
Affiliation(s)
- Chao Hu
- Shanghai Matwings Technology Co., Ltd., Shanghai, 200240, China
- School of Information Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Song Li
- School of Physics and Astronomy and Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Matwings Technology Co., Ltd., Shanghai, 200240, China
| | - Chenxing Yang
- Shanghai Matwings Technology Co., Ltd., Shanghai, 200240, China
| | - Jun Chen
- Shanghai Matwings Technology Co., Ltd., Shanghai, 200240, China
| | - Yi Xiong
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, 201203, China
| | - Guisheng Fan
- School of Information Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Hao Liu
- Shanghai Matwings Technology Co., Ltd., Shanghai, 200240, China.
| | - Liang Hong
- School of Physics and Astronomy and Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, 201203, China.
| |
Collapse
|
14
|
Carullo G, Mazzotta S, Ceramella J, Iacopetta D, Ramunno A, Rosano C, Brizzi A, Campiani G, Aiello F, Sinicropi MS. Development of 1-(2-aminophenyl)pyrrole-based amides acting as human topoisomerase I inhibitors. Arch Pharm (Weinheim) 2023; 356:e2300270. [PMID: 37452410 DOI: 10.1002/ardp.202300270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
Topoisomerases are ubiquitous enzymes in the human body, particularly involved in cancer development and progression. Topoisomerase I (topoI) performs DNA relaxation reactions by "controlled rotation" rather than by "strand passage." The inhibition of topoI has become a useful strategy to control cancer cell proliferation. Nowadays, different compounds have undergone clinical trials, but the search for new molecular entities is necessary and benefits from medicinal chemistry efforts. Pyrrole-based compounds emerged as promising antiproliferative agents, with particular interest in breast cancer therapy and topoI inhibition. Starting from these observations and based on the scaffold-hopping approach, we developed a small library of 1-(2-aminophenyl)pyrrole-based amides (7a-f) as new anticancer agents. Tested on a panel of cancer cell lines, 7a-f displayed the most interesting profile in MDA-MB-231 cells, where the most active compounds, 7d-f, were able to induce death by apoptosis. Direct enzymatic assays and docking simulations on the topoI active site (PDB: 1A35) revealed the inhibitory activity and potential binding site for the newly developed 1-(2-aminophenyl)pyrrole-based amides.
Collapse
Affiliation(s)
- Gabriele Carullo
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Sarah Mazzotta
- Dipartimento di Chimica, Università degli Studi di Milano, Milano, Italy
| | - Jessica Ceramella
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Rende, Italy
| | - Domenico Iacopetta
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Rende, Italy
| | - Anna Ramunno
- Dipartimento di Farmacia, Università degli Studi di Salerno, Fisciano, Italy
| | - Camillo Rosano
- Unità di Proteomica e Spettrometria di Massa, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Antonella Brizzi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Giuseppe Campiani
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Francesca Aiello
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Rende, Italy
| | - Maria S Sinicropi
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Rende, Italy
| |
Collapse
|
15
|
Niazi SK, Mariam Z. Recent Advances in Machine-Learning-Based Chemoinformatics: A Comprehensive Review. Int J Mol Sci 2023; 24:11488. [PMID: 37511247 PMCID: PMC10380192 DOI: 10.3390/ijms241411488] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/30/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
In modern drug discovery, the combination of chemoinformatics and quantitative structure-activity relationship (QSAR) modeling has emerged as a formidable alliance, enabling researchers to harness the vast potential of machine learning (ML) techniques for predictive molecular design and analysis. This review delves into the fundamental aspects of chemoinformatics, elucidating the intricate nature of chemical data and the crucial role of molecular descriptors in unveiling the underlying molecular properties. Molecular descriptors, including 2D fingerprints and topological indices, in conjunction with the structure-activity relationships (SARs), are pivotal in unlocking the pathway to small-molecule drug discovery. Technical intricacies of developing robust ML-QSAR models, including feature selection, model validation, and performance evaluation, are discussed herewith. Various ML algorithms, such as regression analysis and support vector machines, are showcased in the text for their ability to predict and comprehend the relationships between molecular structures and biological activities. This review serves as a comprehensive guide for researchers, providing an understanding of the synergy between chemoinformatics, QSAR, and ML. Due to embracing these cutting-edge technologies, predictive molecular analysis holds promise for expediting the discovery of novel therapeutic agents in the pharmaceutical sciences.
Collapse
Affiliation(s)
- Sarfaraz K Niazi
- College of Pharmacy, University of Illinois, Chicago, IL 61820, USA
| | - Zamara Mariam
- Zamara Mariam, School of Interdisciplinary Engineering & Sciences (SINES), National University of Sciences & Technology (NUST), Islamabad 24090, Pakistan
| |
Collapse
|
16
|
Mokaya M, Imrie F, van Hoorn WP, Kalisz A, Bradley AR, Deane CM. Testing the limits of SMILES-based de novo molecular generation with curriculum and deep reinforcement learning. NAT MACH INTELL 2023. [DOI: 10.1038/s42256-023-00636-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
17
|
Danel T, Łęski J, Podlewska S, Podolak IT. Docking-based generative approaches in the search for new drug candidates. Drug Discov Today 2023; 28:103439. [PMID: 36372330 DOI: 10.1016/j.drudis.2022.103439] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/08/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Despite the popularity of virtual screening (VS) of existing compound libraries, the search for new potential drug candidates also takes advantage of generative protocols, where new compound suggestions are enumerated using various algorithms. To increase the activity potency of generative approaches, they have recently been coupled with molecular docking, a leading methodology of structure-based drug design (SBDD). In this review, we summarize progress since docking-based generative models emerged. We propose a new taxonomy for these methods and discuss their importance for the field of computer-aided drug design (CADD). In addition, we discuss the most promising directions for the further development of generative protocols coupled with docking.
Collapse
Affiliation(s)
- Tomasz Danel
- Faculty of Mathematics and Computer Science, Jagiellonian University, 6 Łojasiewicza Street, 30-348 Kraków, Poland.
| | - Jan Łęski
- Faculty of Mathematics and Computer Science, Jagiellonian University, 6 Łojasiewicza Street, 30-348 Kraków, Poland
| | - Sabina Podlewska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Medicinal Chemistry, 31-343 Kraków, Smętna Street 12, Poland
| | - Igor T Podolak
- Faculty of Mathematics and Computer Science, Jagiellonian University, 6 Łojasiewicza Street, 30-348 Kraków, Poland
| |
Collapse
|
18
|
Abate C, Decherchi S, Cavalli A. Graph neural networks for conditional de novo drug design. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2023. [DOI: 10.1002/wcms.1651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Carlo Abate
- Fondazione Istituto Italiano di Tecnologia Genoa Italy
- Università degli Studi di Bologna Bologna Italy
| | | | - Andrea Cavalli
- Fondazione Istituto Italiano di Tecnologia Genoa Italy
- Università degli Studi di Bologna Bologna Italy
| |
Collapse
|
19
|
Tan Y, Dai L, Huang W, Guo Y, Zheng S, Lei J, Chen H, Yang Y. DRlinker: Deep Reinforcement Learning for Optimization in Fragment Linking Design. J Chem Inf Model 2022; 62:5907-5917. [PMID: 36404642 DOI: 10.1021/acs.jcim.2c00982] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fragment-based drug discovery is a widely used strategy for drug design in both academic and pharmaceutical industries. Although fragments can be linked to generate candidate compounds by the latest deep generative models, generating linkers with specified attributes remains underdeveloped. In this study, we presented a novel framework, DRlinker, to control fragment linking toward compounds with given attributes through reinforcement learning. The method has been shown to be effective for many tasks from controlling the linker length and log P, optimizing predicted bioactivity of compounds, to various multiobjective tasks. Specifically, our model successfully generated 91.0% and 93.9% of compounds complying with the desired linker length and log P and improved the 7.5 pChEMBL value in bioactivity optimization. Finally, a quasi-scaffold-hopping study revealed that DRlinker could generate nearly 30% molecules with high 3D similarity but low 2D similarity to the lead inhibitor, demonstrating the benefits and applicability of DRlinker in actual fragment-based drug design.
Collapse
Affiliation(s)
- Youhai Tan
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou510006, China
| | - Lingxue Dai
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou510006, China
| | - Weifeng Huang
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou510006, China
| | - Yinfeng Guo
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou510006, China
| | - Shuangjia Zheng
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou510006, China.,Galixir Technologies, Beijing100083, China
| | - Jinping Lei
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou510006, China
| | - Hongming Chen
- Guangzhou Laboratory, No. 9 XinDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou510005, China
| | - Yuedong Yang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou510006, China
| |
Collapse
|
20
|
Chandler M, Jain S, Halman J, Hong E, Dobrovolskaia MA, Zakharov AV, Afonin KA. Artificial Immune Cell, AI-cell, a New Tool to Predict Interferon Production by Peripheral Blood Monocytes in Response to Nucleic Acid Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2204941. [PMID: 36216772 PMCID: PMC9671856 DOI: 10.1002/smll.202204941] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/15/2022] [Indexed: 06/16/2023]
Abstract
Nucleic acid nanoparticles, or NANPs, rationally designed to communicate with the human immune system, can offer innovative therapeutic strategies to overcome the limitations of traditional nucleic acid therapies. Each set of NANPs is unique in their architectural parameters and physicochemical properties, which together with the type of delivery vehicles determine the kind and the magnitude of their immune response. Currently, there are no predictive tools that would reliably guide the design of NANPs to the desired immunological outcome, a step crucial for the success of personalized therapies. Through a systematic approach investigating physicochemical and immunological profiles of a comprehensive panel of various NANPs, the research team developes and experimentally validates a computational model based on the transformer architecture able to predict the immune activities of NANPs. It is anticipated that the freely accessible computational tool that is called an "artificial immune cell," or AI-cell, will aid in addressing the current critical public health challenges related to safety criteria of nucleic acid therapies in a timely manner and promote the development of novel biomedical tools.
Collapse
Affiliation(s)
- Morgan Chandler
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Sankalp Jain
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Justin Halman
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Enping Hong
- Nanotechnology Characterization Lab, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Marina A. Dobrovolskaia
- Nanotechnology Characterization Lab, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Alexey V. Zakharov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Kirill A. Afonin
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| |
Collapse
|
21
|
Callis TB, Garrett TR, Montgomery AP, Danon JJ, Kassiou M. Recent Scaffold Hopping Applications in Central Nervous System Drug Discovery. J Med Chem 2022; 65:13483-13504. [PMID: 36206553 DOI: 10.1021/acs.jmedchem.2c00969] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The concept of bioisosterism and the implementation of bioisosteric replacement is fundamental to medicinal chemistry. The exploration of bioisosteres is often used to probe key structural features of candidate pharmacophores and enhance pharmacokinetic properties. As the understanding of bioisosterism has evolved, capabilities to undertake more ambitious bioisosteric replacements have emerged. Scaffold hopping is a broadly used term in the literature referring to a variety of different bioisosteric replacement strategies, ranging from simple heterocyclic replacements to topological structural overhauls. In this work, we have highlighted recent applications of scaffold hopping in the central nervous system drug discovery space. While we have highlighted the benefits of using scaffold hopping approaches in central nervous system drug discovery, these are also widely applicable to other medicinal chemistry fields. We also recommend a shift toward the use of more refined and meaningful terminology within the realm of scaffold hopping.
Collapse
Affiliation(s)
- Timothy B Callis
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Taylor R Garrett
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Jonathan J Danon
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
22
|
Zheng S, Tan Y, Wang Z, Li C, Zhang Z, Sang X, Chen H, Yang Y. Accelerated rational PROTAC design via deep learning and molecular simulations. NAT MACH INTELL 2022. [DOI: 10.1038/s42256-022-00527-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|