1
|
Zhao Y, Yuan C, Shi Y, Liu X, Luo L, Zhang L, Pešić M, Yao H, Li L. Drug screening approaches for small-molecule compounds in cancer-targeted therapy. J Drug Target 2025; 33:368-383. [PMID: 39575843 DOI: 10.1080/1061186x.2024.2427185] [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: 05/30/2024] [Revised: 09/30/2024] [Accepted: 10/27/2024] [Indexed: 02/08/2025]
Abstract
Small-molecule compounds exhibit distinct pharmacological properties and clinical effectiveness. Over the past decade, advances in covalent drug discovery have led to successful small-molecule drugs, such as EGFR, BTK, and KRAS (G12C) inhibitors, for cancer therapy. Researchers are paying more attention to refining drug screening methods aiming for high throughput, fast speed, high specificity, and accuracy. Therefore, the discovery and development of small-molecule drugs has been facilitated by significantly reducing screening time and financial resources, and increasing promising lead compounds compared with traditional methods. This review aims to introduce classical and emerging methods for screening small-molecule compounds in targeted cancer therapy. It includes classification, principles, advantages, disadvantages, and successful applications, serving as valuable references for subsequent researchers.
Collapse
Affiliation(s)
- Yelin Zhao
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenyu Yuan
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuchen Shi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohong Liu
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Xicheng District, Beijing, China
| | - Liaoxin Luo
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Li Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research, 'Siniša Stanković'- National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Hongjuan Yao
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Li
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Srivastava N, Verma S, Singh A, Shukla P, Singh Y, Oza AD, Kaur T, Chowdhury S, Kapoor M, Yadav AN. Advances in artificial intelligence-based technologies for increasing the quality of medical products. Daru 2024; 33:1. [PMID: 39613923 DOI: 10.1007/s40199-024-00548-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 10/09/2024] [Indexed: 12/01/2024] Open
Abstract
Artificial intelligence (AI) is a technology that combines machine learning (ML) and deep learning. It has numerous usages in the domains of medicine and other sciences. Artificial intelligence can forecast the behavior of a drug's target protein and predict its desired physicochemical qualities. AI's potential to enhance healthcare services offerings formerly unheard-of opportunities for cost reserves, enhanced overall clinical and patient outcomes. The recent development of research in the biomedical field, encompassing fields such as genomics, computational medicine, AI, and algorithms for learning, has led to the demand for novel technology, a fresh workforce, and new standards of practice set the stage for the revolution in healthcare. By connecting these health statistics with cutting-edge AI technologies, precise insights into patient treatment can be obtained. Moreover, AI can aid in the search for new drugs by foretelling the target protein's two-dimensional structure. In the current review, an overview of the latest AI-based technologies and how they may be employed to reduce product development time to market and snowballing product quality, cost-effectiveness, as well as security throughout the manufacturing process is detailed.
Collapse
Affiliation(s)
- Nidhi Srivastava
- Maharishi School of Pharmaceutical Sciences, Maharishi University of Information and Technology, Lucknow, Uttar Pradesh, India.
| | - Sneha Verma
- Maharishi School of Science and Humanities, Maharishi University of Information and Technology, Lucknow, Uttar Pradesh, India
| | - Anupama Singh
- Maharishi School of Pharmaceutical Sciences, Maharishi University of Information and Technology, Lucknow, Uttar Pradesh, India
| | - Pranki Shukla
- Maharishi School of Pharmaceutical Sciences, Maharishi University of Information and Technology, Lucknow, Uttar Pradesh, India
| | - Yashvardhan Singh
- Maharishi School of Pharmaceutical Sciences, Maharishi University of Information and Technology, Lucknow, Uttar Pradesh, India
| | - Ankit D Oza
- Department of Mechanical Engineering, Parul Institute of Technology, Parul University, Vadodara, Gujarat, India
| | - Tanvir Kaur
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Sohini Chowdhury
- Chitkara Centre for Research and Development, Chitkara University, Baddi, Himachal Pradesh, India
| | - Monit Kapoor
- Centre of Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Ajar Nath Yadav
- Department of Genetics, Plant Breeding and Biotechnology, Dr. Khem Singh Gill Akal College of Agriculture, Eternal University, Baru Sahib, Sirmour, Himachal Pradesh, India.
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India.
| |
Collapse
|
3
|
Bharadwaj S, Deepika K, Kumar A, Jaiswal S, Miglani S, Singh D, Fartyal P, Kumar R, Singh S, Singh MP, Gaidhane AM, Kumar B, Jha V. Exploring the Artificial Intelligence and Its Impact in Pharmaceutical Sciences: Insights Toward the Horizons Where Technology Meets Tradition. Chem Biol Drug Des 2024; 104:e14639. [PMID: 39396920 DOI: 10.1111/cbdd.14639] [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: 07/27/2024] [Revised: 09/03/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024]
Abstract
The technological revolutions in computers and the advancement of high-throughput screening technologies have driven the application of artificial intelligence (AI) for faster discovery of drug molecules with more efficiency, and cost-friendly finding of hit or lead molecules. The ability of software and network frameworks to interpret molecular structures' representations and establish relationships/correlations has enabled various research teams to develop numerous AI platforms for identifying new lead molecules or discovering new targets for already established drug molecules. The prediction of biological activity, ADME properties, and toxicity parameters in early stages have reduced the chances of failure and associated costs in later clinical stages, which was observed at a high rate in the tedious, expensive, and laborious drug discovery process. This review focuses on the different AI and machine learning (ML) techniques with their applications mainly focused on the pharmaceutical industry. The applications of AI frameworks in the identification of molecular target, hit identification/hit-to-lead optimization, analyzing drug-receptor interactions, drug repurposing, polypharmacology, synthetic accessibility, clinical trial design, and pharmaceutical developments are discussed in detail. We have also compiled the details of various startups in AI in this field. This review will provide a comprehensive analysis and outline various state-of-the-art AI/ML techniques to the readers with their framework applications. This review also highlights the challenges in this field, which need to be addressed for further success in pharmaceutical applications.
Collapse
Affiliation(s)
- Shruti Bharadwaj
- Center for SeNSE, Indian Institute of Technology Delhi (IIT), New Delhi, India
| | - Kumari Deepika
- Department of Computer Engineering, Pune Institute of Computer Technology, Pune, India
| | - Asim Kumar
- Amity Institute of Pharmacy (AIP), Amity University Haryana, Manesar, India
| | - Shivani Jaiswal
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Shaweta Miglani
- Department of Education, Central University of Punjab, Bathinda, India
| | - Damini Singh
- IES Institute of Pharmacy, IES University, Bhopal, Madhya Pradesh, India
| | - Prachi Fartyal
- Department of Mathematics, Govt PG College Bajpur (US Nagar), Bazpur, Uttarakhand, India
| | - Roshan Kumar
- Department of Microbiology, Graphic Era (Deemed to be University), Dehradun, India
- Department of Microbiology, Central University of Punjab, VPO-Ghudda, Punjab, India
| | - Shareen Singh
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Mahendra Pratap Singh
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Abhay M Gaidhane
- Jawaharlal Nehru Medical College, and Global Health Academy, School of Epidemiology and Public Health, Datta Meghe Institute of Higher Education, Wardha, India
| | - Bhupinder Kumar
- Department of Pharmaceutical Science, Hemvati Nandan Bahuguna Garhwal (A Central) University, Srinagar, Uttarakhand, India
| | - Vibhu Jha
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
4
|
Duo L, Liu Y, Ren J, Tang B, Hirst JD. Artificial intelligence for small molecule anticancer drug discovery. Expert Opin Drug Discov 2024; 19:933-948. [PMID: 39074493 DOI: 10.1080/17460441.2024.2367014] [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: 04/22/2024] [Accepted: 06/07/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION The transition from conventional cytotoxic chemotherapy to targeted cancer therapy with small-molecule anticancer drugs has enhanced treatment outcomes. This approach, which now dominates cancer treatment, has its advantages. Despite the regulatory approval of several targeted molecules for clinical use, challenges such as low response rates and drug resistance still persist. Conventional drug discovery methods are costly and time-consuming, necessitating more efficient approaches. The rise of artificial intelligence (AI) and access to large-scale datasets have revolutionized the field of small-molecule cancer drug discovery. Machine learning (ML), particularly deep learning (DL) techniques, enables the rapid identification and development of novel anticancer agents by analyzing vast amounts of genomic, proteomic, and imaging data to uncover hidden patterns and relationships. AREA COVERED In this review, the authors explore the important landmarks in the history of AI-driven drug discovery. They also highlight various applications in small-molecule cancer drug discovery, outline the challenges faced, and provide insights for future research. EXPERT OPINION The advent of big data has allowed AI to penetrate and enable innovations in almost every stage of medicine discovery, transforming the landscape of oncology research through the development of state-of-the-art algorithms and models. Despite challenges in data quality, model interpretability, and technical limitations, advancements promise breakthroughs in personalized and precision oncology, revolutionizing future cancer management.
Collapse
Affiliation(s)
- Lihui Duo
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Yu Liu
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Jianfeng Ren
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Bencan Tang
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Jonathan D Hirst
- School of Chemistry, University of Nottingham University Park, Nottingham, UK
| |
Collapse
|
5
|
Scarano N, Brullo C, Musumeci F, Millo E, Bruzzone S, Schenone S, Cichero E. Recent Advances in the Discovery of SIRT1/2 Inhibitors via Computational Methods: A Perspective. Pharmaceuticals (Basel) 2024; 17:601. [PMID: 38794171 PMCID: PMC11123952 DOI: 10.3390/ph17050601] [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: 03/30/2024] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/26/2024] Open
Abstract
Sirtuins (SIRTs) are classified as class III histone deacetylases (HDACs), a family of enzymes that catalyze the removal of acetyl groups from the ε-N-acetyl lysine residues of histone proteins, thus counteracting the activity performed by histone acetyltransferares (HATs). Based on their involvement in different biological pathways, ranging from transcription to metabolism and genome stability, SIRT dysregulation was investigated in many diseases, such as cancer, neurodegenerative disorders, diabetes, and cardiovascular and autoimmune diseases. The elucidation of a consistent number of SIRT-ligand complexes helped to steer the identification of novel and more selective modulators. Due to the high diversity and quantity of the structural data thus far available, we reviewed some of the different ligands and structure-based methods that have recently been used to identify new promising SIRT1/2 modulators. The present review is structured into two sections: the first includes a comprehensive perspective of the successful computational approaches related to the discovery of SIRT1/2 inhibitors (SIRTIs); the second section deals with the most interesting SIRTIs that have recently appeared in the literature (from 2017). The data reported here are collected from different databases (SciFinder, Web of Science, Scopus, Google Scholar, and PubMed) using "SIRT", "sirtuin", and "sirtuin inhibitors" as keywords.
Collapse
Affiliation(s)
- Naomi Scarano
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (F.M.); (S.S.)
| | - Chiara Brullo
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (F.M.); (S.S.)
| | - Francesca Musumeci
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (F.M.); (S.S.)
| | - Enrico Millo
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy; (E.M.); (S.B.)
| | - Santina Bruzzone
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy; (E.M.); (S.B.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Silvia Schenone
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (F.M.); (S.S.)
| | - Elena Cichero
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (F.M.); (S.S.)
| |
Collapse
|
6
|
Habeeb M, You HW, Umapathi M, Ravikumar KK, Hariyadi, Mishra S. Strategies of Artificial intelligence tools in the domain of nanomedicine. J Drug Deliv Sci Technol 2024; 91:105157. [DOI: 10.1016/j.jddst.2023.105157] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
7
|
Pan S, Xia L, Xu L, Li Z. SubMDTA: drug target affinity prediction based on substructure extraction and multi-scale features. BMC Bioinformatics 2023; 24:334. [PMID: 37679724 PMCID: PMC10485962 DOI: 10.1186/s12859-023-05460-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Drug-target affinity (DTA) prediction is a critical step in the field of drug discovery. In recent years, deep learning-based methods have emerged for DTA prediction. In order to solve the problem of fusion of substructure information of drug molecular graphs and utilize multi-scale information of protein, a self-supervised pre-training model based on substructure extraction and multi-scale features is proposed in this paper. RESULTS For drug molecules, the model obtains substructure information through the method of probability matrix, and the contrastive learning method is implemented on the graph-level representation and subgraph-level representation to pre-train the graph encoder for downstream tasks. For targets, a BiLSTM method that integrates multi-scale features is used to capture long-distance relationships in the amino acid sequence. The experimental results showed that our model achieved better performance for DTA prediction. CONCLUSIONS The proposed model improves the performance of the DTA prediction, which provides a novel strategy based on substructure extraction and multi-scale features.
Collapse
Affiliation(s)
- Shourun Pan
- College of Computer Science and Technology, Qingdao University, Qingdao, China
| | - Leiming Xia
- College of Computer Science and Technology, Qingdao University, Qingdao, China
| | - Lei Xu
- College of Computer Science and Technology, Qingdao University, Qingdao, China
| | - Zhen Li
- College of Computer Science and Technology, Qingdao University, Qingdao, China.
| |
Collapse
|
8
|
Wen J, Gan H, Yang Z, Zhou R, Zhao J, Ye Z. Mutual-DTI: A mutual interaction feature-based neural network for drug-target protein interaction prediction. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:10610-10625. [PMID: 37322951 DOI: 10.3934/mbe.2023469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The prediction of drug-target protein interaction (DTI) is a crucial task in the development of new drugs in modern medicine. Accurately identifying DTI through computer simulations can significantly reduce development time and costs. In recent years, many sequence-based DTI prediction methods have been proposed, and introducing attention mechanisms has improved their forecasting performance. However, these methods have some shortcomings. For example, inappropriate dataset partitioning during data preprocessing can lead to overly optimistic prediction results. Additionally, only single non-covalent intermolecular interactions are considered in the DTI simulation, ignoring the complex interactions between their internal atoms and amino acids. In this paper, we propose a network model called Mutual-DTI that predicts DTI based on the interaction properties of sequences and a Transformer model. We use multi-head attention to extract the long-distance interdependent features of the sequence and introduce a module to extract the sequence's mutual interaction features in mining complex reaction processes of atoms and amino acids. We evaluate the experiments on two benchmark datasets, and the results show that Mutual-DTI outperforms the latest baseline significantly. In addition, we conduct ablation experiments on a label-inversion dataset that is split more rigorously. The results show that there is a significant improvement in the evaluation metrics after introducing the extracted sequence interaction feature module. This suggests that Mutual-DTI may contribute to modern medical drug development research. The experimental results show the effectiveness of our approach. The code for Mutual-DTI can be downloaded from https://github.com/a610lab/Mutual-DTI.
Collapse
Affiliation(s)
- Jiahui Wen
- School of Computer Science, Hubei University of Technology, Wuhan 430068, China
| | - Haitao Gan
- School of Computer Science, Hubei University of Technology, Wuhan 430068, China
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan 430062, China
| | - Zhi Yang
- School of Computer Science, Hubei University of Technology, Wuhan 430068, China
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan 430062, China
| | - Ran Zhou
- School of Computer Science, Hubei University of Technology, Wuhan 430068, China
| | - Jing Zhao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan 430062, China
| | - Zhiwei Ye
- School of Computer Science, Hubei University of Technology, Wuhan 430068, China
| |
Collapse
|
9
|
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
|
10
|
Hua Y, Song X, Feng Z, Wu XJ, Kittler J, Yu DJ. CPInformer for Efficient and Robust Compound-Protein Interaction Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:285-296. [PMID: 35044921 DOI: 10.1109/tcbb.2022.3144008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Recently, deep learning has become the mainstream methodology for Compound-Protein Interaction (CPI) prediction. However, the existing compound-protein feature extraction methods have some issues that limit their performance. First, graph networks are widely used for structural compound feature extraction, but the chemical properties of a compound depend on functional groups rather than graphic structure. Besides, the existing methods lack capabilities in extracting rich and discriminative protein features. Last, the compound-protein features are usually simply combined for CPI prediction, without considering information redundancy and effective feature mining. To address the above issues, we propose a novel CPInformer method. Specifically, we extract heterogeneous compound features, including structural graph features and functional class fingerprints, to reduce prediction errors caused by similar structural compounds. Then, we combine local and global features using dense connections to obtain multi-scale protein features. Last, we apply ProbSparse self-attention to protein features, under the guidance of compound features, to eliminate information redundancy, and to improve the accuracy of CPInformer. More importantly, the proposed method identifies the activated local regions that link a CPI, providing a good visualisation for the CPI state. The results obtained on five benchmarks demonstrate the merits and superiority of CPInformer over the state-of-the-art approaches.
Collapse
|
11
|
Cheng Z, Zhao Q, Li Y, Wang J. IIFDTI: predicting drug-target interactions through interactive and independent features based on attention mechanism. Bioinformatics 2022; 38:4153-4161. [PMID: 35801934 DOI: 10.1093/bioinformatics/btac485] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/02/2022] [Accepted: 07/07/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Identifying drug-target interactions is a crucial step for drug discovery and design. Traditional biochemical experiments are credible to accurately validate drug-target interactions. However, they are also extremely laborious, time-consuming and expensive. With the collection of more validated biomedical data and the advancement of computing technology, the computational methods based on chemogenomics gradually attract more attention, which guide the experimental verifications. RESULTS In this study, we propose an end-to-end deep learning-based method named IIFDTI to predict drug-target interactions (DTIs) based on independent features of drug-target pairs and interactive features of their substructures. First, the interactive features of substructures between drugs and targets are extracted by the bidirectional encoder-decoder architecture. The independent features of drugs and targets are extracted by the graph neural networks and convolutional neural networks, respectively. Then, all extracted features are fused and inputted into fully connected dense layers in downstream tasks for predicting DTIs. IIFDTI takes into account the independent features of drugs/targets and simulates the interactive features of the substructures from the biological perspective. Multiple experiments show that IIFDTI outperforms the state-of-the-art methods in terms of the area under the receiver operating characteristics curve (AUC), the area under the precision-recall curve (AUPR), precision, and recall on benchmark datasets. In addition, the mapped visualizations of attention weights indicate that IIFDTI has learned the biological knowledge insights, and two case studies illustrate the capabilities of IIFDTI in practical applications. AVAILABILITY AND IMPLEMENTATION The data and codes underlying this article are available in Github at https://github.com/czjczj/IIFDTI. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Zhongjian Cheng
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Qichang Zhao
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Yaohang Li
- Department of Computer Science, Old Dominion University, Norfolk, VA 23529, USA
| | - Jianxin Wang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
12
|
Abbotto E, Scarano N, Piacente F, Millo E, Cichero E, Bruzzone S. Virtual Screening in the Identification of Sirtuins’ Activity Modulators. Molecules 2022; 27:molecules27175641. [PMID: 36080416 PMCID: PMC9457788 DOI: 10.3390/molecules27175641] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Sirtuins are NAD+-dependent deac(et)ylases with different subcellular localization. The sirtuins’ family is composed of seven members, named SIRT-1 to SIRT-7. Their substrates include histones and also an increasing number of different proteins. Sirtuins regulate a wide range of different processes, ranging from transcription to metabolism to genome stability. Thus, their dysregulation has been related to the pathogenesis of different diseases. In this review, we discussed the pharmacological approaches based on sirtuins’ modulators (both inhibitors and activators) that have been attempted in in vitro and/or in in vivo experimental settings, to highlight the therapeutic potential of targeting one/more specific sirtuin isoform(s) in cancer, neurodegenerative disorders and type 2 diabetes. Extensive research has already been performed to identify SIRT-1 and -2 modulators, while compounds targeting the other sirtuins have been less studied so far. Beside sections dedicated to each sirtuin, in the present review we also included sections dedicated to pan-sirtuins’ and to parasitic sirtuins’ modulators. A special focus is dedicated to the sirtuins’ modulators identified by the use of virtual screening.
Collapse
Affiliation(s)
- Elena Abbotto
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
| | - Naomi Scarano
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Francesco Piacente
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
| | - Enrico Millo
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
| | - Elena Cichero
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
- Correspondence:
| |
Collapse
|
13
|
ZHANG BY, ZHENG YF, ZHAO J, KANG D, WANG Z, XU LJ, LIU AL, DU GH. Identification of multi-target anti-cancer agents from TCM formula by in silico prediction and in vitro validation. Chin J Nat Med 2022; 20:332-351. [DOI: 10.1016/s1875-5364(22)60180-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Indexed: 11/03/2022]
|
14
|
Sahoo BM, Bhattamisra SK, Das S, Tiwari A, Tiwari V, Kumar M, Singh S. Computational Approach to Combat COVID-19 Infection: Emerging Tool for Accelerating Drug Research. Curr Drug Discov Technol 2022; 19:e170122200314. [PMID: 35040405 DOI: 10.2174/1570163819666220117161308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/05/2021] [Accepted: 10/11/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Drug discovery and development process is an expensive, complex, time-consuming and risky. There are different techniques involved in the drug development process which include random screening, computational approach, molecular manipulation and serendipitous research. Among these methods, the computational approach is considered as an efficient strategy to accelerate and economize the drug discovery process. OBJECTIVE This approach is mainly applied in various phases of drug discovery process including target identification, target validation, lead identification and lead optimization. Due to increase in the availability of information regarding various biological targets of different disease states, computational approaches such as molecular docking, de novo design, molecular similarity calculation, virtual screening, pharmacophore-based modeling and pharmacophore mapping have been applied extensively. METHODS Various drug molecules can be designed by applying computational tools to explore the drug candidates for treatment of Coronavirus infection. The world health organization has announced the novel corona virus disease as COVID-19 and declared it as pandemic globally on 11 February 2020. So, it is thought of interest to scientific community to apply computational methods to design and optimize the pharmacological properties of various clinically available and FDA approved drugs such as remdesivir, ribavirin, favipiravir, oseltamivir, ritonavir, arbidol, chloroquine, hydroxychloroquine, carfilzomib, baraticinib, prulifloxacin, etc for effective treatment of COVID-19 infection. RESULTS Further, various survey reports suggest that the extensive studies are carried out by various research communities to find out the safety and efficacy profile of these drug candidates. CONCLUSION This review is focused on the study of various aspects of these drugs related to their target sites on virus, binding interactions, physicochemical properties etc.
Collapse
Affiliation(s)
- Biswa Mohan Sahoo
- Roland Institute of Pharmaceutical Sciences, Berhampur-760010, Odisha, India
| | - Subrat Kumar Bhattamisra
- Department of Pharmaceutical Technology, School of Medical Sciences, Adamas University, Jagannathpur, Kolkata-700126, West Bengal, India
| | - Sarita Das
- Microbiology Laboratory, Department of Botany, Berhampur University, Bhanja Bihar, Berhampur- 760007, Odisha, India
| | - Abhishek Tiwari
- Devasthali Vidyapeeth College of Pharmacy, Lalpur, Rudrapur-263148, Uttarakhand, India
| | - Varsha Tiwari
- Devasthali Vidyapeeth College of Pharmacy, Lalpur, Rudrapur-263148, Uttarakhand, India
| | - Manish Kumar
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala-133207, Haryana, India
| | - Sunil Singh
- Shri Sai College of Pharmacy, Handia, Prayagraj, Uttar Pradesh, 221503, India
| |
Collapse
|
15
|
Jin Y, Lu J, Shi R, Yang Y. EmbedDTI: Enhancing the Molecular Representations via Sequence Embedding and Graph Convolutional Network for the Prediction of Drug-Target Interaction. Biomolecules 2021; 11:biom11121783. [PMID: 34944427 PMCID: PMC8698792 DOI: 10.3390/biom11121783] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/20/2021] [Accepted: 11/24/2021] [Indexed: 01/09/2023] Open
Abstract
The identification of drug-target interaction (DTI) plays a key role in drug discovery and development. Benefitting from large-scale drug databases and verified DTI relationships, a lot of machine-learning methods have been developed to predict DTIs. However, due to the difficulty in extracting useful information from molecules, the performance of these methods is limited by the representation of drugs and target proteins. This study proposes a new model called EmbedDTI to enhance the representation of both drugs and target proteins, and improve the performance of DTI prediction. For protein sequences, we leverage language modeling for pretraining the feature embeddings of amino acids and feed them to a convolutional neural network model for further representation learning. For drugs, we build two levels of graphs to represent compound structural information, namely the atom graph and substructure graph, and adopt graph convolutional network with an attention module to learn the embedding vectors for the graphs. We compare EmbedDTI with the existing DTI predictors on two benchmark datasets. The experimental results show that EmbedDTI outperforms the state-of-the-art models, and the attention module can identify the components crucial for DTIs in compounds.
Collapse
Affiliation(s)
- Yuan Jin
- Center for Brain-Like Computing and Machine Intelligence, Department of Computer Science and Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Rd., Shanghai 200240, China; (Y.J.); (R.S.)
| | - Jiarui Lu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Rd., Shanghai 200240, China;
| | - Runhan Shi
- Center for Brain-Like Computing and Machine Intelligence, Department of Computer Science and Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Rd., Shanghai 200240, China; (Y.J.); (R.S.)
| | - Yang Yang
- Center for Brain-Like Computing and Machine Intelligence, Department of Computer Science and Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Rd., Shanghai 200240, China; (Y.J.); (R.S.)
- Key Laboratory of Shanghai Education Commission for Intelligent Interaction and Cognitive Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Rd., Shanghai 200240, China
- Correspondence:
| |
Collapse
|
16
|
He S, Leanse LG, Feng Y. Artificial intelligence and machine learning assisted drug delivery for effective treatment of infectious diseases. Adv Drug Deliv Rev 2021; 178:113922. [PMID: 34461198 DOI: 10.1016/j.addr.2021.113922] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/14/2021] [Accepted: 08/09/2021] [Indexed: 12/23/2022]
Abstract
In the era of antimicrobial resistance, the prevalence of multidrug-resistant microorganisms that resist conventional antibiotic treatment has steadily increased. Thus, it is now unquestionable that infectious diseases are significant global burdens that urgently require innovative treatment strategies. Emerging studies have demonstrated that artificial intelligence (AI) can transform drug delivery to promote effective treatment of infectious diseases. In this review, we propose to evaluate the significance, essential principles, and popular tools of AI in drug delivery for infectious disease treatment. Specifically, we will focus on the achievements and key findings of current research, as well as the applications of AI on drug delivery throughout the whole antimicrobial treatment process, with an emphasis on drug development, treatment regimen optimization, drug delivery system and administration route design, and drug delivery outcome prediction. To that end, the challenges of AI in drug delivery for infectious disease treatments and their current solutions and future perspective will be presented and discussed.
Collapse
Affiliation(s)
- Sheng He
- Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, MA, USA.
| | - Leon G Leanse
- Massachusetts General Hospital, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Yanfang Feng
- Massachusetts General Hospital, Harvard Medical School, Harvard University, Boston, MA, USA.
| |
Collapse
|
17
|
Paul D, Sanap G, Shenoy S, Kalyane D, Kalia K, Tekade RK. Artificial intelligence in drug discovery and development. Drug Discov Today 2021; 26:80-93. [PMID: 33099022 PMCID: PMC7577280 DOI: 10.1016/j.drudis.2020.10.010] [Citation(s) in RCA: 432] [Impact Index Per Article: 108.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/03/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023]
Abstract
Artificial intelligence-integrated drug discovery and development has accelerated the growth of the pharmaceutical sector, leading to a revolutionary change in the pharma industry. Here, we discuss areas of integration, tools, and techniques utilized in enforcing AI, ongoing challenges, and ways to overcome them.
Collapse
Affiliation(s)
- Debleena Paul
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India
| | - Gaurav Sanap
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India
| | - Snehal Shenoy
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India
| | - Dnyaneshwar Kalyane
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India
| | - Kiran Kalia
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India
| | - Rakesh K Tekade
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
18
|
Vlachakis D, Vlamos P. Mathematical Multidimensional Modelling and Structural Artificial Intelligence Pipelines Provide Insights for the Designing of Highly Specific AntiSARS-CoV2 Agents. MATHEMATICS IN COMPUTER SCIENCE 2021; 15. [PMCID: PMC8205651 DOI: 10.1007/s11786-021-00517-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
COVID19 is the most impactful pandemic of recent times worldwide. It is a highly infectious disease that is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 virus), To date there is specific drug nor vaccination against COVID19. Therefor the need for novel and pioneering anti-COVID19 is of paramount importance. In this direction, computer-aided drug design constitutes a very promising antiviral approach for the discovery and analysis of drugs and molecules with biological activity against SARS-CoV2. In silico modelling takes advantage of the massive amounts of biological and chemical data available on the nature of the interactions between the targeted systems and molecules, as well as the rapid progress of computational tools and software. Herein, we describe the potential of the merging of mathematical modelling, artificial intelligence and learning techniques into seamless computational pipelines for the rapid and efficient discovery and design of potent anti- SARS-CoV-2 modulators.
Collapse
Affiliation(s)
- Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, Genetics and Computational Biology Group, School of Applied Biology and Biotechnology, Agricultural University of Athens, Iera Odos 75 Str. GR11855, Athens, Greece
- Laboratory of Molecular Endocrinology, Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Soranou Ephessiou Str. GR11527, Athens, Greece
- University Research Institute of Maternal and Child Health and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Thivon 1 & Papadiamantopoulou Str. GR11527, Athens, Greece
| | - Panayiotis Vlamos
- Department of Informatics, Ionian University, Plateia Tsirigoti 7, 49100 Corfu, Greece
| |
Collapse
|
19
|
Chen L, Tan X, Wang D, Zhong F, Liu X, Yang T, Luo X, Chen K, Jiang H, Zheng M. TransformerCPI: improving compound–protein interaction prediction by sequence-based deep learning with self-attention mechanism and label reversal experiments. Bioinformatics 2020; 36:4406-4414. [DOI: 10.1093/bioinformatics/btaa524] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/13/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022] Open
Abstract
Abstract
Motivation
Identifying compound–protein interaction (CPI) is a crucial task in drug discovery and chemogenomics studies, and proteins without three-dimensional structure account for a large part of potential biological targets, which requires developing methods using only protein sequence information to predict CPI. However, sequence-based CPI models may face some specific pitfalls, including using inappropriate datasets, hidden ligand bias and splitting datasets inappropriately, resulting in overestimation of their prediction performance.
Results
To address these issues, we here constructed new datasets specific for CPI prediction, proposed a novel transformer neural network named TransformerCPI, and introduced a more rigorous label reversal experiment to test whether a model learns true interaction features. TransformerCPI achieved much improved performance on the new experiments, and it can be deconvolved to highlight important interacting regions of protein sequences and compound atoms, which may contribute chemical biology studies with useful guidance for further ligand structural optimization.
Availability and implementation
https://github.com/lifanchen-simm/transformerCPI.
Collapse
Affiliation(s)
- Lifan Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoqin Tan
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dingyan Wang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feisheng Zhong
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaohong Liu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Tianbiao Yang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaomin Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kaixian Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Hualiang Jiang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
20
|
Lee I, Keum J, Nam H. DeepConv-DTI: Prediction of drug-target interactions via deep learning with convolution on protein sequences. PLoS Comput Biol 2019; 15:e1007129. [PMID: 31199797 PMCID: PMC6594651 DOI: 10.1371/journal.pcbi.1007129] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 06/26/2019] [Accepted: 05/24/2019] [Indexed: 12/04/2022] Open
Abstract
Identification of drug-target interactions (DTIs) plays a key role in drug discovery. The high cost and labor-intensive nature of in vitro and in vivo experiments have highlighted the importance of in silico-based DTI prediction approaches. In several computational models, conventional protein descriptors have been shown to not be sufficiently informative to predict accurate DTIs. Thus, in this study, we propose a deep learning based DTI prediction model capturing local residue patterns of proteins participating in DTIs. When we employ a convolutional neural network (CNN) on raw protein sequences, we perform convolution on various lengths of amino acids subsequences to capture local residue patterns of generalized protein classes. We train our model with large-scale DTI information and demonstrate the performance of the proposed model using an independent dataset that is not seen during the training phase. As a result, our model performs better than previous protein descriptor-based models. Also, our model performs better than the recently developed deep learning models for massive prediction of DTIs. By examining pooled convolution results, we confirmed that our model can detect binding sites of proteins for DTIs. In conclusion, our prediction model for detecting local residue patterns of target proteins successfully enriches the protein features of a raw protein sequence, yielding better prediction results than previous approaches. Our code is available at https://github.com/GIST-CSBL/DeepConv-DTI.
Collapse
Affiliation(s)
- Ingoo Lee
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Buk-ku, Gwangju, Republic of Korea
| | - Jongsoo Keum
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Buk-ku, Gwangju, Republic of Korea
| | - Hojung Nam
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Buk-ku, Gwangju, Republic of Korea
| |
Collapse
|
21
|
Network pharmacology-based analysis of Chinese herbal Naodesheng formula for application to Alzheimer's disease. Chin J Nat Med 2018; 16:53-62. [PMID: 29425590 DOI: 10.1016/s1875-5364(18)30029-3] [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: 05/08/2017] [Indexed: 12/13/2022]
Abstract
Naodesheng (NDS) formula, which consists of Rhizoma Chuanxiong, Lobed Kudzuvine, Carthamus tinctorius, Radix Notoginseng, and Crataegus pinnatifida, is widely applied for the treatment of cardio/cerebrovascular ischemic diseases, ischemic stroke, and sequelae of cerebral hemorrhage, etc. At present, the studies on NDS formula for Alzheimer's disease (AD) only focus on single component of this prescription, and there is no report about the synergistic mechanism of the constituents in NDS formula for the potential treatment of dementia. Therefore, the present study aimed to predict the potential targets and uncover the mechanisms of NDS formula for the treatment of AD. Firstly, we collected the constituents in NDS formula and key targets toward AD. Then, drug-likeness, oral bioavailability, and blood-brain barrier permeability were evaluated to find drug-like and lead-like constituents for treatment of central nervous system diseases. By combining the advantages of machine learning, molecular docking, and pharmacophore mapping, we attempted to predict the targets of constituents and find potential multi-target compounds from NDS formula. Finally, we built constituent-target network, constituent-target-target network and target-biological pathway network to study the network pharmacology of the constituents in NDS formula. To the best of our knowledge, this represented the first to study the mechanism of NDS formula for potential efficacy for AD treatment by means of the virtual screening and network pharmacology methods.
Collapse
|
22
|
Abstract
Aim: Computational chemogenomics models the compound–protein interaction space, typically for drug discovery, where existing methods predominantly either incorporate increasing numbers of bioactivity samples or focus on specific subfamilies of proteins and ligands. As an alternative to modeling entire large datasets at once, active learning adaptively incorporates a minimum of informative examples for modeling, yielding compact but high quality models. Results/methodology: We assessed active learning for protein/target family-wide chemogenomic modeling by replicate experiment. Results demonstrate that small yet highly predictive models can be extracted from only 10–25% of large bioactivity datasets, irrespective of molecule descriptors used. Conclusion: Chemogenomic active learning identifies small subsets of ligand–target interactions in a large screening database that lead to knowledge discovery and highly predictive models.
Collapse
|
23
|
Abstract
Drug discovery is a multidisciplinary and multivariate optimization endeavor. As such, in silico screening tools have gained considerable importance to archive, analyze and exploit the vast and ever-increasing amount of experimental data generated throughout the process. The current review will focus on the computer-aided prediction of the numerous properties that need to be controlled during the discovery of a preliminary hit and its promotion to a viable clinical candidate. It does not pretend to the almost impossible task of an exhaustive report but will highlight a few key points that need to be collectively addressed both by chemists and biologists to fuel the drug discovery pipeline with innovative and safe drug candidates.
Collapse
Affiliation(s)
- Didier Rognan
- Laboratoire d'Innovation Thérapeutique, UMR 7200 CNRS-Université de Strasbourg, 74 route du Rhin, 67400 Illkirch, France.
| |
Collapse
|
24
|
HajiEbrahimi A, Ghafouri H, Ranjbar M, Sakhteman A. Protein Ligand Interaction Fingerprints. PHARMACEUTICAL SCIENCES 2017. [DOI: 10.4018/978-1-5225-1762-7.ch041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A most challenging part in docking-based virtual screening is the scoring functions implemented in various docking programs in order to evaluate different poses of the ligands inside the binding cavity of the receptor. Precise and trustable measurement of ligand-protein affinity for Structure-Based Virtual Screening (SB-VS) is therefore, an outstanding problem in docking studies. Empirical post-docking filters can be helpful as a way to provide various types of structure-activity information. Different types of interaction have been presented between the ligands and the receptor so far. Based on the diversity and importance of PLIF methods, this chapter will focus on the comparison of different protocols. The advantages and disadvantages of all methods will be discussed explicitly in this chapter as well as future sights for further progress in this field. Different classifications approaches for the protein-ligand interaction fingerprints were also discussed in this chapter.
Collapse
|
25
|
Tian K, Shao M, Wang Y, Guan J, Zhou S. Boosting compound-protein interaction prediction by deep learning. Methods 2016; 110:64-72. [PMID: 27378654 DOI: 10.1016/j.ymeth.2016.06.024] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/20/2016] [Accepted: 06/28/2016] [Indexed: 11/19/2022] Open
Abstract
The identification of interactions between compounds and proteins plays an important role in network pharmacology and drug discovery. However, experimentally identifying compound-protein interactions (CPIs) is generally expensive and time-consuming, computational approaches are thus introduced. Among these, machine-learning based methods have achieved a considerable success. However, due to the nonlinear and imbalanced nature of biological data, many machine learning approaches have their own limitations. Recently, deep learning techniques show advantages over many state-of-the-art machine learning methods in some applications. In this study, we aim at improving the performance of CPI prediction based on deep learning, and propose a method called DL-CPI (the abbreviation of Deep Learning for Compound-Protein Interactions prediction), which employs deep neural network (DNN) to effectively learn the representations of compound-protein pairs. Extensive experiments show that DL-CPI can learn useful features of compound-protein pairs by a layerwise abstraction, and thus achieves better prediction performance than existing methods on both balanced and imbalanced datasets.
Collapse
Affiliation(s)
- Kai Tian
- Shanghai Key Lab of Intelligent Information Processing, School of Computer Science, Fudan University, Shanghai 200433, China.
| | - Mingyu Shao
- Shanghai Key Lab of Intelligent Information Processing, School of Computer Science, Fudan University, Shanghai 200433, China.
| | - Yang Wang
- School of Software, Jiangxi Normal University, Nanchang 330022, China.
| | - Jihong Guan
- Department of Computer Science and Technology, Tongji University, Shanghai 201804, China.
| | - Shuigeng Zhou
- Shanghai Key Lab of Intelligent Information Processing, School of Computer Science, Fudan University, Shanghai 200433, China.
| |
Collapse
|
26
|
Chiba S, Ikeda K, Ishida T, Gromiha MM, Taguchi YH, Iwadate M, Umeyama H, Hsin KY, Kitano H, Yamamoto K, Sugaya N, Kato K, Okuno T, Chikenji G, Mochizuki M, Yasuo N, Yoshino R, Yanagisawa K, Ban T, Teramoto R, Ramakrishnan C, Thangakani AM, Velmurugan D, Prathipati P, Ito J, Tsuchiya Y, Mizuguchi K, Honma T, Hirokawa T, Akiyama Y, Sekijima M. Identification of potential inhibitors based on compound proposal contest: Tyrosine-protein kinase Yes as a target. Sci Rep 2015; 5:17209. [PMID: 26607293 PMCID: PMC4660442 DOI: 10.1038/srep17209] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/27/2015] [Indexed: 12/14/2022] Open
Abstract
A search of broader range of chemical space is important for drug discovery. Different methods of computer-aided drug discovery (CADD) are known to propose compounds in different chemical spaces as hit molecules for the same target protein. This study aimed at using multiple CADD methods through open innovation to achieve a level of hit molecule diversity that is not achievable with any particular single method. We held a compound proposal contest, in which multiple research groups participated and predicted inhibitors of tyrosine-protein kinase Yes. This showed whether collective knowledge based on individual approaches helped to obtain hit compounds from a broad range of chemical space and whether the contest-based approach was effective.
Collapse
Affiliation(s)
- Shuntaro Chiba
- Education Academy of Computational Life Sciences (ACLS), Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama 226-8501 Japan
| | - Kazuyoshi Ikeda
- Level Five Co. Ltd., Shiodome Shibarikyu Bldg., 1-2-3 Kaigan, Minato-ku, Tokyo 105-0022, Japan
| | - Takashi Ishida
- Education Academy of Computational Life Sciences (ACLS), Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama 226-8501 Japan.,Department of Computer Science, Tokyo Institute of Technology, 2-12-1, Ookayama, Meguro-ku, Tokyo 152-8550 Japan
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600 036, Tamilnadu, India
| | - Y-H Taguchi
- Department of Physics, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Mitsuo Iwadate
- Department of Biological Sciences, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Hideaki Umeyama
- Department of Biological Sciences, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Kun-Yi Hsin
- Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami, Okinawa 904-0495 Japan
| | - Hiroaki Kitano
- Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami, Okinawa 904-0495 Japan.,The Systems Biology Research Institute, Falcon Building 5F, 5-6-9 Shirokanedai, Minato-ku, Tokyo 108-0071 Japan.,Center for Integrative Medical Sciences, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
| | - Kazuki Yamamoto
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904 Japan
| | - Nobuyoshi Sugaya
- PharmaDesign Inc., 2-19-8, Hatchobori, Chuo-ku, Tokyo 104-0032 Japan
| | - Koya Kato
- Department of Computational Science and Engineering, Nagoya University, Furocho, Chikusa, Nagoya 464-8603, Japan
| | - Tatsuya Okuno
- Division of Neurogenetics, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - George Chikenji
- Department of Computational Science and Engineering, Nagoya University, Furocho, Chikusa, Nagoya 464-8603, Japan
| | - Masahiro Mochizuki
- Information and Mathematical Science and Bioinformatics Co., Ltd., Level 6 OWL TOWER, 4-21-1 Higashi-Ikebukuro, Toshima-ku, Tokyo 170-0013 Japan
| | - Nobuaki Yasuo
- Education Academy of Computational Life Sciences (ACLS), Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama 226-8501 Japan.,Department of Computer Science, Tokyo Institute of Technology, 2-12-1, Ookayama, Meguro-ku, Tokyo 152-8550 Japan
| | - Ryunosuke Yoshino
- Global Scientific Information and Computing Center, Tokyo Institute of Technology 2-12-1, Ookayama, Meguro-ku, Tokyo 152-8550 Japan.,Department of Biotechnology, The University of Tokyo, 1-1-1 Yayoi, Nunkyo-ku, Tokyo, 113-8657
| | - Keisuke Yanagisawa
- Education Academy of Computational Life Sciences (ACLS), Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama 226-8501 Japan.,Department of Computer Science, Tokyo Institute of Technology, 2-12-1, Ookayama, Meguro-ku, Tokyo 152-8550 Japan
| | - Tomohiro Ban
- Education Academy of Computational Life Sciences (ACLS), Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama 226-8501 Japan.,Department of Computer Science, Tokyo Institute of Technology, 2-12-1, Ookayama, Meguro-ku, Tokyo 152-8550 Japan
| | - Reiji Teramoto
- Forerunner Pharma Research, Co., Ltd., Yokohama Bio Industry Center, 1-6 Shuehiro-cho, Tsurumi-ku, Yokohama 230-0045 Japan
| | - Chandrasekaran Ramakrishnan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600 036, Tamilnadu, India
| | - A Mary Thangakani
- Centre of Advanced Study in Crystallography and Biophysics and Bioinformatics Infrastructure Facility (DBT Funded), University of Madras, Chennai 600025, Tamilnadu, India
| | - D Velmurugan
- Centre of Advanced Study in Crystallography and Biophysics and Bioinformatics Infrastructure Facility (DBT Funded), University of Madras, Chennai 600025, Tamilnadu, India
| | - Philip Prathipati
- National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085 Japan
| | - Junichi Ito
- National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085 Japan
| | - Yuko Tsuchiya
- National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085 Japan
| | - Kenji Mizuguchi
- National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085 Japan
| | - Teruki Honma
- Center for Life Science Technologies, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe-shi, Hyogo 650-0047 Japan
| | - Takatsugu Hirokawa
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo, 135-0064, Japan.,Initiative for Parallel Bioinformatics, Level 14 Hibiya Central Building, 1-2-9 Nishi-Shimbashi Minato-Ku, Tokyo 105-0003 Japan
| | - Yutaka Akiyama
- Education Academy of Computational Life Sciences (ACLS), Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama 226-8501 Japan.,Department of Computer Science, Tokyo Institute of Technology, 2-12-1, Ookayama, Meguro-ku, Tokyo 152-8550 Japan.,Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo, 135-0064, Japan.,Initiative for Parallel Bioinformatics, Level 14 Hibiya Central Building, 1-2-9 Nishi-Shimbashi Minato-Ku, Tokyo 105-0003 Japan
| | - Masakazu Sekijima
- Education Academy of Computational Life Sciences (ACLS), Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama 226-8501 Japan.,Department of Computer Science, Tokyo Institute of Technology, 2-12-1, Ookayama, Meguro-ku, Tokyo 152-8550 Japan.,Global Scientific Information and Computing Center, Tokyo Institute of Technology 2-12-1, Ookayama, Meguro-ku, Tokyo 152-8550 Japan.,Initiative for Parallel Bioinformatics, Level 14 Hibiya Central Building, 1-2-9 Nishi-Shimbashi Minato-Ku, Tokyo 105-0003 Japan
| |
Collapse
|
27
|
Developing a QSAR model for hepatotoxicity screening of the active compounds in traditional Chinese medicines. Food Chem Toxicol 2015; 78:71-7. [DOI: 10.1016/j.fct.2015.01.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 01/10/2023]
|
28
|
Jasial S, Balfer J, Vogt M, Bajorath J. Determination of Meta-Parameters for Support Vector Machine Linear Combinations. Mol Inform 2015; 34:127-33. [DOI: 10.1002/minf.201400163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/16/2014] [Indexed: 11/05/2022]
|
29
|
Fang J, Li Y, Liu R, Pang X, Li C, Yang R, He Y, Lian W, Liu AL, Du GH. Discovery of multitarget-directed ligands against Alzheimer's disease through systematic prediction of chemical-protein interactions. J Chem Inf Model 2015; 55:149-64. [PMID: 25531792 DOI: 10.1021/ci500574n] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
To determine chemical-protein interactions (CPI) is costly, time-consuming, and labor-intensive. In silico prediction of CPI can facilitate the target identification and drug discovery. Although many in silico target prediction tools have been developed, few of them could predict active molecules against multitarget for a single disease. In this investigation, naive Bayesian (NB) and recursive partitioning (RP) algorithms were applied to construct classifiers for predicting the active molecules against 25 key targets toward Alzheimer's disease (AD) using the multitarget-quantitative structure-activity relationships (mt-QSAR) method. Each molecule was initially represented with two kinds of fingerprint descriptors (ECFP6 and MACCS). One hundred classifiers were constructed, and their performance was evaluated and verified with internally 5-fold cross-validation and external test set validation. The range of the area under the receiver operating characteristic curve (ROC) for the test sets was from 0.741 to 1.0, with an average of 0.965. In addition, the important fragments for multitarget against AD given by NB classifiers were also analyzed. Finally, the validated models were employed to systematically predict the potential targets for six approved anti-AD drugs and 19 known active compounds related to AD. The prediction results were confirmed by reported bioactivity data and our in vitro experimental validation, resulting in several multitarget-directed ligands (MTDLs) against AD, including seven acetylcholinesterase (AChE) inhibitors ranging from 0.442 to 72.26 μM and four histamine receptor 3 (H3R) antagonists ranging from 0.308 to 58.6 μM. To be exciting, the best MTDL DL0410 was identified as an dual cholinesterase inhibitor with IC50 values of 0.442 μM (AChE) and 3.57 μM (BuChE) as well as a H3R antagonist with an IC50 of 0.308 μM. This investigation is the first report using mt-QASR approach to predict chemical-protein interaction for a single disease and discovering highly potent MTDLs. This protocol may be useful for in silico multitarget prediction of other diseases.
Collapse
Affiliation(s)
- Jiansong Fang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing 100050, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Quaternary ammonium and amido derivatives of pyranochromenones and chromenones: synthesis and antimicrobial activity evaluation. Med Chem Res 2014. [DOI: 10.1007/s00044-014-1294-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
31
|
Korkmaz S, Zararsiz G, Goksuluk D. Drug/nondrug classification using Support Vector Machines with various feature selection strategies. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2014; 117:51-60. [PMID: 25224081 DOI: 10.1016/j.cmpb.2014.08.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 08/15/2014] [Accepted: 08/27/2014] [Indexed: 06/03/2023]
Abstract
In conjunction with the advance in computer technology, virtual screening of small molecules has been started to use in drug discovery. Since there are thousands of compounds in early-phase of drug discovery, a fast classification method, which can distinguish between active and inactive molecules, can be used for screening large compound collections. In this study, we used Support Vector Machines (SVM) for this type of classification task. SVM is a powerful classification tool that is becoming increasingly popular in various machine-learning applications. The data sets consist of 631 compounds for training set and 216 compounds for a separate test set. In data pre-processing step, the Pearson's correlation coefficient used as a filter to eliminate redundant features. After application of the correlation filter, a single SVM has been applied to this reduced data set. Moreover, we have investigated the performance of SVM with different feature selection strategies, including SVM-Recursive Feature Elimination, Wrapper Method and Subset Selection. All feature selection methods generally represent better performance than a single SVM while Subset Selection outperforms other feature selection methods. We have tested SVM as a classification tool in a real-life drug discovery problem and our results revealed that it could be a useful method for classification task in early-phase of drug discovery.
Collapse
Affiliation(s)
- Selcuk Korkmaz
- Hacettepe University, Faculty of Medicine, Department of Biostatistics, 06100 Sihhiye, Ankara, Turkey.
| | - Gokmen Zararsiz
- Hacettepe University, Faculty of Medicine, Department of Biostatistics, 06100 Sihhiye, Ankara, Turkey
| | - Dincer Goksuluk
- Hacettepe University, Faculty of Medicine, Department of Biostatistics, 06100 Sihhiye, Ankara, Turkey
| |
Collapse
|
32
|
Sugaya N. Ligand efficiency-based support vector regression models for predicting bioactivities of ligands to drug target proteins. J Chem Inf Model 2014; 54:2751-63. [PMID: 25220713 DOI: 10.1021/ci5003262] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The concept of ligand efficiency (LE) indices is widely accepted throughout the drug design community and is frequently used in a retrospective manner in the process of drug development. For example, LE indices are used to investigate LE optimization processes of already-approved drugs and to re-evaluate hit compounds obtained from structure-based virtual screening methods and/or high-throughput experimental assays. However, LE indices could also be applied in a prospective manner to explore drug candidates. Here, we describe the construction of machine learning-based regression models in which LE indices are adopted as an end point and show that LE-based regression models can outperform regression models based on pIC50 values. In addition to pIC50 values traditionally used in machine learning studies based on chemogenomics data, three representative LE indices (ligand lipophilicity efficiency (LLE), binding efficiency index (BEI), and surface efficiency index (SEI)) were adopted, then used to create four types of training data. We constructed regression models by applying a support vector regression (SVR) method to the training data. In cross-validation tests of the SVR models, the LE-based SVR models showed higher correlations between the observed and predicted values than the pIC50-based models. Application tests to new data displayed that, generally, the predictive performance of SVR models follows the order SEI > BEI > LLE > pIC50. Close examination of the distributions of the activity values (pIC50, LLE, BEI, and SEI) in the training and validation data implied that the performance order of the SVR models may be ascribed to the much higher diversity of the LE-based training and validation data. In the application tests, the LE-based SVR models can offer better predictive performance of compound-protein pairs with a wider range of ligand potencies than the pIC50-based models. This finding strongly suggests that LE-based SVR models are better than pIC50-based models at predicting bioactivities of compounds that could exhibit a much higher (or lower) potency.
Collapse
Affiliation(s)
- Nobuyoshi Sugaya
- Drug Discovery Department, Research & Development Division, PharmaDesign, Inc. , Hatchobori 2-19-8, Chuo-ku, Tokyo 104-0032, Japan
| |
Collapse
|
33
|
Liu X, Xu Y, Li S, Wang Y, Peng J, Luo C, Luo X, Zheng M, Chen K, Jiang H. In Silico target fishing: addressing a "Big Data" problem by ligand-based similarity rankings with data fusion. J Cheminform 2014; 6:33. [PMID: 24976868 PMCID: PMC4068908 DOI: 10.1186/1758-2946-6-33] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 06/10/2014] [Indexed: 11/16/2022] Open
Abstract
Background Ligand-based in silico target fishing can be used to identify the potential interacting target of bioactive ligands, which is useful for understanding the polypharmacology and safety profile of existing drugs. The underlying principle of the approach is that known bioactive ligands can be used as reference to predict the targets for a new compound. Results We tested a pipeline enabling large-scale target fishing and drug repositioning, based on simple fingerprint similarity rankings with data fusion. A large library containing 533 drug relevant targets with 179,807 active ligands was compiled, where each target was defined by its ligand set. For a given query molecule, its target profile is generated by similarity searching against the ligand sets assigned to each target, for which individual searches utilizing multiple reference structures are then fused into a single ranking list representing the potential target interaction profile of the query compound. The proposed approach was validated by 10-fold cross validation and two external tests using data from DrugBank and Therapeutic Target Database (TTD). The use of the approach was further demonstrated with some examples concerning the drug repositioning and drug side-effects prediction. The promising results suggest that the proposed method is useful for not only finding promiscuous drugs for their new usages, but also predicting some important toxic liabilities. Conclusions With the rapid increasing volume and diversity of data concerning drug related targets and their ligands, the simple ligand-based target fishing approach would play an important role in assisting future drug design and discovery.
Collapse
Affiliation(s)
- Xian Liu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yuan Xu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Shanshan Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yulan Wang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Jianlong Peng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Cheng Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Xiaomin Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Kaixian Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China ; School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Hualiang Jiang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China ; School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| |
Collapse
|
34
|
Li S, Zhang J, Lu S, Huang W, Geng L, Shen Q, Zhang J. The mechanism of allosteric inhibition of protein tyrosine phosphatase 1B. PLoS One 2014; 9:e97668. [PMID: 24831294 PMCID: PMC4022711 DOI: 10.1371/journal.pone.0097668] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 04/09/2014] [Indexed: 11/24/2022] Open
Abstract
As the prototypical member of the PTP family, protein tyrosine phosphatase 1B (PTP1B) is an attractive target for therapeutic interventions in type 2 diabetes. The extremely conserved catalytic site of PTP1B renders the design of selective PTP1B inhibitors intractable. Although discovered allosteric inhibitors containing a benzofuran sulfonamide scaffold offer fascinating opportunities to overcome selectivity issues, the allosteric inhibitory mechanism of PTP1B has remained elusive. Here, molecular dynamics (MD) simulations, coupled with a dynamic weighted community analysis, were performed to unveil the potential allosteric signal propagation pathway from the allosteric site to the catalytic site in PTP1B. This result revealed that the allosteric inhibitor compound-3 induces a conformational rearrangement in helix α7, disrupting the triangular interaction among helix α7, helix α3, and loop11. Helix α7 then produces a force, pulling helix α3 outward, and promotes Ser190 to interact with Tyr176. As a result, the deviation of Tyr176 abrogates the hydrophobic interactions with Trp179 and leads to the downward movement of the WPD loop, which forms an H-bond between Asp181 and Glu115. The formation of this H-bond constrains the WPD loop to its open conformation and thus inactivates PTP1B. The discovery of this allosteric mechanism provides an overall view of the regulation of PTP1B, which is an important insight for the design of potent allosteric PTP1B inhibitors.
Collapse
Affiliation(s)
- Shuai Li
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai JiaoTong University, School of Medicine (SJTU-SM), Shanghai, China
| | - Jingmiao Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai JiaoTong University, School of Medicine (SJTU-SM), Shanghai, China
| | - Shaoyong Lu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai JiaoTong University, School of Medicine (SJTU-SM), Shanghai, China
| | - Wenkang Huang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai JiaoTong University, School of Medicine (SJTU-SM), Shanghai, China
| | - Lv Geng
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai JiaoTong University, School of Medicine (SJTU-SM), Shanghai, China
| | - Qiancheng Shen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai JiaoTong University, School of Medicine (SJTU-SM), Shanghai, China
| | - Jian Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai JiaoTong University, School of Medicine (SJTU-SM), Shanghai, China
- Medicinal Bioinformatics Center, Shanghai JiaoTong University, School of Medicine, Shanghai, China
| |
Collapse
|
35
|
Abstract
Allostery is the most direct and efficient way for regulation of biological macromolecule function, ranging from the control of metabolic mechanisms to signal transduction pathways. Allosteric modulators target to allosteric sites, offering distinct advantages compared to orthosteric ligands that target to active sites, such as greater specificity, reduced side effects, and lower toxicity. Allosteric modulators have therefore drawn increasing attention as potential therapeutic drugs in the design and development of new drugs. In recent years, advancements in our understanding of the fundamental principles underlying allostery, coupled with the exploitation of powerful techniques and methods in the field of allostery, provide unprecedented opportunities to discover allosteric proteins, detect and characterize allosteric sites, design and develop novel efficient allosteric drugs, and recapitulate the universal features of allosteric proteins and allosteric modulators. In the present review, we summarize the recent advances in the repertoire of allostery, with a particular focus on the aforementioned allosteric compounds.
Collapse
Affiliation(s)
- Shaoyong Lu
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai, China
| | | | | |
Collapse
|
36
|
Fechner P, Bleher O, Ewald M, Freudenberger K, Furin D, Hilbig U, Kolarov F, Krieg K, Leidner L, Markovic G, Proll G, Pröll F, Rau S, Riedt J, Schwarz B, Weber P, Widmaier J. Size does matter! Label-free detection of small molecule-protein interaction. Anal Bioanal Chem 2014; 406:4033-51. [PMID: 24817356 DOI: 10.1007/s00216-014-7834-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/07/2014] [Accepted: 04/11/2014] [Indexed: 11/28/2022]
Abstract
This review is focused on methods for detecting small molecules and, in particular, the characterisation of their interaction with natural proteins (e.g. receptors, ion channels). Because there are intrinsic advantages to using label-free methods over labelled methods (e.g. fluorescence, radioactivity), this review only covers label-free techniques. We briefly discuss available techniques and their advantages and disadvantages, especially as related to investigating the interaction between small molecules and proteins. The reviewed techniques include well-known and widely used standard analytical methods (e.g. HPLC-MS, NMR, calorimetry, and X-ray diffraction), newer and more specialised analytical methods (e.g. biosensors), biological systems (e.g. cell lines and animal models), and in-silico approaches.
Collapse
Affiliation(s)
- Peter Fechner
- Biametrics GmbH, Auf der Morgenstelle 18, 72076, Tübingen, Germany,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
|
39
|
Sugaya N. Training based on ligand efficiency improves prediction of bioactivities of ligands and drug target proteins in a machine learning approach. J Chem Inf Model 2013; 53:2525-37. [PMID: 24020509 DOI: 10.1021/ci400240u] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Machine learning methods based on ligand-protein interaction data in bioactivity databases are one of the current strategies for efficiently finding novel lead compounds as the first step in the drug discovery process. Although previous machine learning studies have succeeded in predicting novel ligand-protein interactions with high performance, all of the previous studies to date have been heavily dependent on the simple use of raw bioactivity data of ligand potencies measured by IC50, EC50, K(i), and K(d) deposited in databases. ChEMBL provides us with a unique opportunity to investigate whether a machine-learning-based classifier created by reflecting ligand efficiency other than the IC50, EC50, K(i), and Kd values can also offer high predictive performance. Here we report that classifiers created from training data based on ligand efficiency show higher performance than those from data based on IC50 or K(i) values. Utilizing GPCRSARfari and KinaseSARfari databases in ChEMBL, we created IC50- or K(i)-based training data and binding efficiency index (BEI) based training data then constructed classifiers using support vector machines (SVMs). The SVM classifiers from the BEI-based training data showed slightly higher area under curve (AUC), accuracy, sensitivity, and specificity in the cross-validation tests. Application of the classifiers to the validation data demonstrated that the AUCs and specificities of the BEI-based classifiers dramatically increased in comparison with the IC50- or K(i)-based classifiers. The improvement of the predictive power by the BEI-based classifiers can be attributed to (i) the more separated distributions of positives and negatives, (ii) the higher diversity of negatives in the BEI-based training data in a feature space of SVMs, and (iii) a more balanced number of positives and negatives in the BEI-based training data. These results strongly suggest that training data based on ligand efficiency as well as data based on classical IC50, EC50, K(d), and K(i) values are important when creating a classifier using a machine learning approach based on bioactivity data.
Collapse
Affiliation(s)
- Nobuyoshi Sugaya
- Drug Discovery Department, Research & Development Division, PharmaDesign, Inc. , Hatchobori 2-19-8, Chuo-ku, Tokyo, 104-0032, Japan
| |
Collapse
|
40
|
Rognan D. Towards the Next Generation of Computational Chemogenomics Tools. Mol Inform 2013; 32:1029-34. [PMID: 27481148 DOI: 10.1002/minf.201300054] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 06/11/2013] [Indexed: 01/07/2023]
Affiliation(s)
- D Rognan
- UMR 7200 CNRS-Université de Strasbourg, MEDALIS Drug Discovery Center, 74 route du Rhin, 67400, Illkirch, France.
| |
Collapse
|
41
|
Medina-Franco JL, Aguayo-Ortiz R. Progress in the Visualization and Mining of Chemical and Target Spaces. Mol Inform 2013; 32:942-53. [DOI: 10.1002/minf.201300041] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 05/06/2013] [Indexed: 01/15/2023]
|
42
|
Niu B, Zhang Y, Ding J, Lu Y, Wang M, Lu W, Yuan X, Yin J. Predicting network of drug-enzyme interaction based on machine learning method. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1844:214-23. [PMID: 23907006 DOI: 10.1016/j.bbapap.2013.07.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 07/16/2013] [Accepted: 07/18/2013] [Indexed: 12/11/2022]
Abstract
It is important to correctly and efficiently map drugs and enzymes to their possible interaction network in modern drug research. In this work, a novel approach was introduced to encode drug and enzyme molecules with physicochemical molecular descriptors and pseudo amino acid composition, respectively. Based on this encoding method, Random Forest was adopted to build the drug-enzyme interaction network. After selecting the optimal features that are able to represent the main factors of drug-enzyme interaction in our prediction, a total of 129 features were attained which can be clustered into nine categories: Elemental Analysis, Geometry, Chemistry, Amino Acid Composition, Secondary Structure, Polarity, Molecular Volume, Codon Diversity and Electrostatic Charge. It is further found that Geometry features were the most important of all the features. As a result, our predicting model achieved an MCC of 0.915 and a sensitivity of 87.9% at the specificity level of 99.8% for 10-fold cross-validation test, and achieved an MCC of 0.895 and a sensitivity of 95.7% at the specificity level of 95.4% for independent set test. This article is part of a Special Issue entitled: Computational Proteomics, Systems Biology & Clinical Implications. Guest Editor: Yudong Cai.
Collapse
Affiliation(s)
- Bing Niu
- College of Life Science, Shanghai University, 99 Shang-Da Road, Shanghai 200072, China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Cao DS, Zhou GH, Liu S, Zhang LX, Xu QS, He M, Liang YZ. Large-scale prediction of human kinase-inhibitor interactions using protein sequences and molecular topological structures. Anal Chim Acta 2013; 792:10-8. [PMID: 23910962 DOI: 10.1016/j.aca.2013.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 07/03/2013] [Accepted: 07/04/2013] [Indexed: 01/28/2023]
Abstract
The kinase family is one of the largest target families in the human genome. The family's key function in signal transduction for all organisms makes it a very attractive target class for the therapeutic interventions in many diseases states such as cancer, diabetes, inflammation and arthritis. A first step toward accelerating kinase drug discovery process is to fast identify whether a chemical and a kinase interact or not. Experimentally, these interactions can be identified by in vitro binding assay - an expensive and laborious procedure that is not applicable on a large scale. Therefore, there is an urgent need to develop statistically efficient approaches for identifying kinase-inhibitor interactions. For the first time, the quantitative binding affinities of kinase-inhibitor pairs are differentiated as a measurement to define if an inhibitor interacts with a kinase, and then a chemogenomics framework using an unbiased set of general integrated features (drug descriptors and protein descriptors) and random forest (RF) is employed to construct a predictive model which can accurately classify kinase-inhibitor pairs. Our results show that RF with integrated features gave prediction accuracy of 93.76%, sensitivity of 92.26%, and specificity of 95.27%, respectively. The results are superior to those by only considering two separated spaces (chemical space and protein space), demonstrating that these integrated features contribute cooperatively. Based on the constructed model, we provided a high confidence list of drug-target associations for subsequent experimental investigation guidance at a low false discovery rate.
Collapse
Affiliation(s)
- Dong-Sheng Cao
- School of Pharmaceutical Sciences, Central South University, Changsha 410013, PR China.
| | | | | | | | | | | | | |
Collapse
|
44
|
Cheng F, Li W, Wu Z, Wang X, Zhang C, Li J, Liu G, Tang Y. Prediction of polypharmacological profiles of drugs by the integration of chemical, side effect, and therapeutic space. J Chem Inf Model 2013; 53:753-62. [PMID: 23527559 DOI: 10.1021/ci400010x] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Prediction of polypharmacological profiles of drugs enables us to investigate drug side effects and further find their new indications, i.e. drug repositioning, which could reduce the costs while increase the productivity of drug discovery. Here we describe a new computational framework to predict polypharmacological profiles of drugs by the integration of chemical, side effect, and therapeutic space. On the basis of our previous developed drug side effects database, named MetaADEDB, a drug side effect similarity inference (DSESI) method was developed for drug-target interaction (DTI) prediction on a known DTI network connecting 621 approved drugs and 893 target proteins. The area under the receiver operating characteristic curve was 0.882 ± 0.011 averaged from 100 simulated tests of 10-fold cross-validation for the DSESI method, which is comparative with drug structural similarity inference and drug therapeutic similarity inference methods. Seven new predicted candidate target proteins for seven approved drugs were confirmed by published experiments, with the successful hit rate more than 15.9%. Moreover, network visualization of drug-target interactions and off-target side effect associations provide new mechanism-of-action of three approved antipsychotic drugs in a case study. The results indicated that the proposed methods could be helpful for prediction of polypharmacological profiles of drugs.
Collapse
Affiliation(s)
- Feixiong Cheng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Cao DS, Liang YZ, Deng Z, Hu QN, He M, Xu QS, Zhou GH, Zhang LX, Deng ZX, Liu S. Genome-scale screening of drug-target associations relevant to Ki using a chemogenomics approach. PLoS One 2013; 8:e57680. [PMID: 23577055 PMCID: PMC3618265 DOI: 10.1371/journal.pone.0057680] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 01/27/2013] [Indexed: 11/18/2022] Open
Abstract
The identification of interactions between drugs and target proteins plays a key role in genomic drug discovery. In the present study, the quantitative binding affinities of drug-target pairs are differentiated as a measurement to define whether a drug interacts with a protein or not, and then a chemogenomics framework using an unbiased set of general integrated features and random forest (RF) is employed to construct a predictive model which can accurately classify drug-target pairs. The predictability of the model is further investigated and validated by several independent validation sets. The built model is used to predict drug-target associations, some of which were confirmed by comparing experimental data from public biological resources. A drug-target interaction network with high confidence drug-target pairs was also reconstructed. This network provides further insight for the action of drugs and targets. Finally, a web-based server called PreDPI-Ki was developed to predict drug-target interactions for drug discovery. In addition to providing a high-confidence list of drug-target associations for subsequent experimental investigation guidance, these results also contribute to the understanding of drug-target interactions. We can also see that quantitative information of drug-target associations could greatly promote the development of more accurate models. The PreDPI-Ki server is freely available via: http://sdd.whu.edu.cn/dpiki.
Collapse
Affiliation(s)
- Dong-Sheng Cao
- Research Center of Modernization of Traditional Chinese Medicines, Central South University, Changsha, P. R. China
| | - Yi-Zeng Liang
- Research Center of Modernization of Traditional Chinese Medicines, Central South University, Changsha, P. R. China
- * E-mail: (YZL); (QNH)
| | - Zhe Deng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, P. R. China
| | - Qian-Nan Hu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, P. R. China
- * E-mail: (YZL); (QNH)
| | - Min He
- Research Center of Modernization of Traditional Chinese Medicines, Central South University, Changsha, P. R. China
| | - Qing-Song Xu
- School of Mathematics and Statistics, Central South University, Changsha, P. R. China
| | - Guang-Hua Zhou
- The 163rd Hospital of The Chinese People's Liberation Army, Changsha, P. R. China
| | - Liu-Xia Zhang
- The 163rd Hospital of The Chinese People's Liberation Army, Changsha, P. R. China
| | - Zi-xin Deng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, P. R. China
| | - Shao Liu
- Xiangya Hospital, Central South University, Changsha, P. R. China
| |
Collapse
|
46
|
Desaphy J, Raimbaud E, Ducrot P, Rognan D. Encoding protein-ligand interaction patterns in fingerprints and graphs. J Chem Inf Model 2013; 53:623-37. [PMID: 23432543 DOI: 10.1021/ci300566n] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We herewith present a novel and universal method to convert protein-ligand coordinates into a simple fingerprint of 210 integers registering the corresponding molecular interaction pattern. Each interaction (hydrophobic, aromatic, hydrogen bond, ionic bond, metal complexation) is detected on the fly and physically described by a pseudoatom centered either on the interacting ligand atom, the interacting protein atom, or the geometric center of both interacting atoms. Counting all possible triplets of interaction pseudoatoms within six distance ranges, and pruning the full integer vector to keep the most frequent triplets enables the definition of a simple (210 integers) and coordinate frame-invariant interaction pattern descriptor (TIFP) that can be applied to compare any pair of protein-ligand complexes. TIFP fingerprints have been calculated for ca. 10,000 druggable protein-ligand complexes therefore enabling a wide comparison of relationships between interaction pattern similarity and ligand or binding site pairwise similarity. We notably show that interaction pattern similarity strongly depends on binding site similarity. In addition to the TIFP fingerprint which registers intermolecular interactions between a ligand and its target protein, we developed two tools (Ishape, Grim) to align protein-ligand complexes from their interaction patterns. Ishape is based on the overlap of interaction pseudoatoms using a smooth Gaussian function, whereas Grim utilizes a standard clique detection algorithm to match interaction pattern graphs. Both tools are complementary and enable protein-ligand complex alignments capitalizing on both global and local pattern similarities. The new fingerprint and companion alignment tools have been successfully used in three scenarios: (i) interaction-biased alignment of protein-ligand complexes, (ii) postprocessing docking poses according to known interaction patterns for a particular target, and (iii) virtual screening for bioisosteric scaffolds sharing similar interaction patterns.
Collapse
Affiliation(s)
- Jérémy Desaphy
- Laboratory for Therapeutical Innovation, UMR 7200 Université de Strabsourg/CNRS , MEDALIS Drug Discovery Center, F-67400 Illkirch, France
| | | | | | | |
Collapse
|
47
|
Ou-Yang SS, Lu JY, Kong XQ, Liang ZJ, Luo C, Jiang H. Computational drug discovery. Acta Pharmacol Sin 2012; 33:1131-40. [PMID: 22922346 PMCID: PMC4003107 DOI: 10.1038/aps.2012.109] [Citation(s) in RCA: 172] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 07/08/2012] [Indexed: 01/09/2023] Open
Abstract
Computational drug discovery is an effective strategy for accelerating and economizing drug discovery and development process. Because of the dramatic increase in the availability of biological macromolecule and small molecule information, the applicability of computational drug discovery has been extended and broadly applied to nearly every stage in the drug discovery and development workflow, including target identification and validation, lead discovery and optimization and preclinical tests. Over the past decades, computational drug discovery methods such as molecular docking, pharmacophore modeling and mapping, de novo design, molecular similarity calculation and sequence-based virtual screening have been greatly improved. In this review, we present an overview of these important computational methods, platforms and successful applications in this field.
Collapse
Affiliation(s)
- Si-sheng Ou-Yang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jun-yan Lu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiang-qian Kong
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhong-jie Liang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Cheng Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hualiang Jiang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
48
|
Vogt M, Bajorath J. Chemoinformatics: A view of the field and current trends in method development. Bioorg Med Chem 2012; 20:5317-23. [DOI: 10.1016/j.bmc.2012.03.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 03/09/2012] [Accepted: 03/12/2012] [Indexed: 12/18/2022]
|
49
|
Prediction of chemical-protein interactions network with weighted network-based inference method. PLoS One 2012; 7:e41064. [PMID: 22815915 PMCID: PMC3397956 DOI: 10.1371/journal.pone.0041064] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 06/16/2012] [Indexed: 12/11/2022] Open
Abstract
Chemical-protein interaction (CPI) is the central topic of target identification and drug discovery. However, large scale determination of CPI is a big challenge for in vitro or in vivo experiments, while in silico prediction shows great advantages due to low cost and high accuracy. On the basis of our previous drug-target interaction prediction via network-based inference (NBI) method, we further developed node- and edge-weighted NBI methods for CPI prediction here. Two comprehensive CPI bipartite networks extracted from ChEMBL database were used to evaluate the methods, one containing 17,111 CPI pairs between 4,741 compounds and 97 G protein-coupled receptors, the other including 13,648 CPI pairs between 2,827 compounds and 206 kinases. The range of the area under receiver operating characteristic curves was 0.73 to 0.83 for the external validation sets, which confirmed the reliability of the prediction. The weak-interaction hypothesis in CPI network was identified by the edge-weighted NBI method. Moreover, to validate the methods, several candidate targets were predicted for five approved drugs, namely imatinib, dasatinib, sertindole, olanzapine and ziprasidone. The molecular hypotheses and experimental evidence for these predictions were further provided. These results confirmed that our methods have potential values in understanding molecular basis of drug polypharmacology and would be helpful for drug repositioning.
Collapse
|
50
|
Cheng F, Zhou Y, Li J, Li W, Liu G, Tang Y. Prediction of chemical-protein interactions: multitarget-QSAR versus computational chemogenomic methods. MOLECULAR BIOSYSTEMS 2012; 8:2373-84. [PMID: 22751809 DOI: 10.1039/c2mb25110h] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Elucidation of chemical-protein interactions (CPI) is the basis of target identification and drug discovery. It is time-consuming and costly to determine CPI experimentally, and computational methods will facilitate the determination of CPI. In this study, two methods, multitarget quantitative structure-activity relationship (mt-QSAR) and computational chemogenomics, were developed for CPI prediction. Two comprehensive data sets were collected from the ChEMBL database for method assessment. One data set consisted of 81 689 CPI pairs among 50 924 compounds and 136 G-protein coupled receptors (GPCRs), while the other one contained 43 965 CPI pairs among 23 376 compounds and 176 kinases. The range of the area under the receiver operating characteristic curve (AUC) for the test sets was 0.95 to 1.0 and 0.82 to 1.0 for 100 GPCR mt-QSAR models and 100 kinase mt-QSAR models, respectively. The AUC of 5-fold cross validation were about 0.92 for both 176 kinases and 136 GPCRs using the chemogenomic method. However, the performance of the chemogenomic method was worse than that of mt-QSAR for the external validation set. Further analysis revealed that there was a high false positive rate for the external validation set when using the chemogenomic method. In addition, we developed a web server named CPI-Predictor, , which is available for free. The methods and tool have potential applications in network pharmacology and drug repositioning.
Collapse
Affiliation(s)
- Feixiong Cheng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | | | | | | | | | | |
Collapse
|