1
|
Feng D, Liu B, Chen Z, Xu J, Geng M, Duan W, Ai J, Zhang H. Discovery of hematopoietic progenitor kinase 1 inhibitors using machine learning-based screening and free energy perturbation. J Biomol Struct Dyn 2025; 43:4152-4164. [PMID: 38198294 DOI: 10.1080/07391102.2024.2301754] [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: 09/12/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is a key negative regulator of T-cell receptor (TCR) signaling and a promising target for cancer immunotherapy. The development of novel HPK1 inhibitors is challenging yet promising. In this study, we used a combination of machine learning (ML)-based virtual screening and free energy perturbation (FEP) calculations to identify novel HPK1 inhibitors. ML-based screening yielded 10 potent HPK1 inhibitors (IC50 < 1 μM). The FEP-guided modification of the in-house false-positive hit, DW21302, revealed that a single key atom change could trigger activity cliffs. The resulting DW21302-A was a potent HPK1 inhibitor (IC50 = 2.1 nM) and potently inhibited cellular HPK1 signaling and enhanced T-cell function. Molecular dynamics (MD) simulations and ADME predictions confirmed DW21302-A as candidate compound. This study provides new strategies and chemical scaffolds for HPK1 inhibitor development.
Collapse
Affiliation(s)
- Dazhi Feng
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Bo Liu
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| | - Zhiwei Chen
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Meiyu Geng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Wenhu Duan
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Jing Ai
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hefeng Zhang
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
2
|
Kumar P, Chaudhary B, Arya P, Chauhan R, Devi S, Parejiya PB, Gupta MM. Advanced Artificial Intelligence Technologies Transforming Contemporary Pharmaceutical Research. Bioengineering (Basel) 2025; 12:363. [PMID: 40281723 PMCID: PMC12024664 DOI: 10.3390/bioengineering12040363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 04/29/2025] Open
Abstract
One area of study within machine learning and artificial intelligence (AI) seeks to create computer programs with intelligence that can mimic human focal processes in order to produce results. This technique includes data collection, effective data usage system development, conclusion illustration, and arrangements. Analysis algorithms that are learning to mimic human cognitive activities are the most widespread application of AI. Artificial intelligence (AI) studies have proliferated, and the field is quickly beginning to understand its potential impact on medical services and investigation. This review delves deeper into the pros and cons of AI across the healthcare and pharmaceutical research industries. Research and review articles published throughout the last few years were selected from PubMed, Google Scholar, and Science Direct, using search terms like 'artificial intelligence', 'drug discovery', 'pharmacy research', 'clinical trial', etc. This article provides a comprehensive overview of how artificial intelligence (AI) is being used to diagnose diseases, treat patients digitally, find new drugs, and predict when outbreaks or pandemics may occur. In artificial intelligence, neural networks and deep learning are some of the most popular tools; in clinical research, Bayesian non-parametric approaches hold promise for better results, while smartphones and the processing of natural languages are employed in recognizing patients and trial monitoring. Seasonal flu, Ebola, Zika, COVID-19, tuberculosis, and outbreak predictions were made using deep computation and artificial intelligence. The academic world is hopeful that AI development will lead to more efficient and less expensive medical and pharmaceutical investigations and better public services.
Collapse
Affiliation(s)
- Parveen Kumar
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University, Jaipur 303121, Rajasthan, India;
| | - Benu Chaudhary
- Shri Ram College of Pharmacy, Karnal 132001, Haryana, India; (B.C.); (P.A.)
| | - Preeti Arya
- Shri Ram College of Pharmacy, Karnal 132001, Haryana, India; (B.C.); (P.A.)
| | - Rupali Chauhan
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (R.C.); (S.D.)
| | - Sushma Devi
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (R.C.); (S.D.)
| | - Punit B. Parejiya
- Department of Pharmaceutics, K.B. Institute of Pharmaceutical Education and Research, Kadi Sarva Vishwavidyalaya, Gandhinagar 382 023, Gujarat, India;
| | - Madan Mohan Gupta
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University, Jaipur 303121, Rajasthan, India;
| |
Collapse
|
3
|
Lateef Junaid MA. Artificial intelligence driven innovations in biochemistry: A review of emerging research frontiers. BIOMOLECULES & BIOMEDICINE 2025; 25:739-750. [PMID: 39819459 PMCID: PMC11959397 DOI: 10.17305/bb.2024.11537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/15/2024] [Accepted: 12/15/2024] [Indexed: 01/19/2025]
Abstract
Artificial intelligence (AI) has become a powerful tool in biochemistry, greatly enhancing research capabilities by enabling the analysis of complex datasets, predicting molecular interactions, and accelerating drug discovery. As AI continues to evolve, its applications in biochemistry are poised to expand, revolutionizing both theoretical and applied research. This review explores current and potential AI applications in biochemistry, with a focus on data analysis, molecular modeling, enzyme engineering, and metabolic pathway studies. Key AI techniques-such as machine learning algorithms, natural language processing, and AI-based molecular modeling-are discussed. The review also highlights emerging research areas benefiting from AI, including personalized medicine and synthetic biology. The methodology involves an extensive analysis of existing literature, particularly peer-reviewed studies on AI applications in biochemistry. AI-driven tools like AlphaFold, which have significantly advanced protein structure prediction, are evaluated alongside AI's role in expediting drug discovery. The review also addresses challenges such as data quality, model interpretability, and ethical considerations. Results indicate that AI has expanded the scope of biochemical research by facilitating large-scale data analysis, enhancing molecular simulations, and opening new avenues of inquiry. However, challenges remain, particularly in data handling and ethical concerns. In conclusion, AI is transforming biochemistry by driving innovation and expanding research possibilities. Future advancements in AI algorithms, interdisciplinary collaboration, and integration with automated techniques will be crucial to fully unlocking AI's potential in advancing biochemical research.
Collapse
Affiliation(s)
- Mohammed Abdul Lateef Junaid
- Department of Basic Medical Sciences, College of Medicine, Majmaah University, Al Majmaah, Kingdom of Saudi Arabia
| |
Collapse
|
4
|
Kim K, Kim J, Kim M, Lee H, Song G. Therapeutic gene target prediction using novel deep hypergraph representation learning. Brief Bioinform 2024; 26:bbaf019. [PMID: 39841592 PMCID: PMC11752618 DOI: 10.1093/bib/bbaf019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/18/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
Identifying therapeutic genes is crucial for developing treatments targeting genetic causes of diseases, but experimental trials are costly and time-consuming. Although many deep learning approaches aim to identify biomarker genes, predicting therapeutic target genes remains challenging due to the limited number of known targets. To address this, we propose HIT (Hypergraph Interaction Transformer), a deep hypergraph representation learning model that identifies a gene's therapeutic potential, biomarker status, or lack of association with diseases. HIT uses hypergraph structures of genes, ontologies, diseases, and phenotypes, employing attention-based learning to capture complex relationships. Experiments demonstrate HIT's state-of-the-art performance, explainability, and ability to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Kibeom Kim
- Division of Artificial Intelligence, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, South Korea
| | - Juseong Kim
- Division of Artificial Intelligence, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, South Korea
| | - Minwook Kim
- Division of Artificial Intelligence, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, South Korea
| | - Hyewon Lee
- Department of Cardiology, Medical Research Institute, Pusan National University Hospital, 179 Gudeok-ro, Busan 49241, South Korea
- College of Medicine, Pusan National University, 20 Geumo-ro, Yangsan 50612, Gyeongsangnam-do, South Korea
| | - Giltae Song
- Division of Artificial Intelligence, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, South Korea
- Department of Electrical and Computer Engineering, School of Computer Science and Engineering, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, South Korea
- Center for Artificial Intelligence Research, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, South Korea
| |
Collapse
|
5
|
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
|
6
|
Varadharajan V, Balu AK, Shiju A, Muthuramalingam P, Shin H, Venkidasamy B, Alharbi NS, Kadaikunnan S, Thiruvengadam M. Deciphering the Anticancer Arsenal of Piper longum: Network Pharmacology and Molecular Docking Unveil Phytochemical Targets Against Lung Cancer. Int J Med Sci 2024; 21:1915-1928. [PMID: 39113883 PMCID: PMC11302554 DOI: 10.7150/ijms.98393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction: Lung cancer, characterized by uncontrolled cellular proliferation within the lung tissues, is the predominant cause of cancer-related fatalities worldwide. The traditional medicinal herb Piper longum has emerged as a significant contender in oncological research because of its documented anticancer attributes, suggesting its potential for novel therapeutic development. Methods: This study adopted network pharmacology and omics methodology to elucidate the anti-lung cancer potential of P. longum by identifying its bioactive constituents and their corresponding molecular targets. Results: Through a comprehensive literature review and the Integrated Medicinal Plant Phytochemistry and Therapeutics database (IMPPAT), we identified 33 bioactive molecules from P. longum. Subsequent analyses employing tools such as SwissTargetPrediction, SuperPred, and DIGEP-Pred facilitated the isolation of 676 potential targets, among which 72 intersected with 666 lung cancer-associated genetic markers identified through databases including the Therapeutic Target Database (TTD), Online Mendelian Inheritance in Man (OMIM), and GeneCards. Further validation through protein-protein interaction (PPI) networks, gene ontology, pathway analyses, boxplots, and overall survival metrics underscored the therapeutic potential of compounds such as 7-epi-eudesm-4(15)-ene-1β, demethoxypiplartine, methyl 3,4,5-trimethoxycinnamate, 6-alpha-diol, and aristolodione. Notably, our findings reaffirm the relevance of lung cancer genes, such as CTNNB1, STAT3, HIF1A, HSP90AA1, and ERBB2, integral to various cellular processes and pivotal in cancer genesis and advancement. Molecular docking assessments revealed pronounced affinity between 6-alpha-diol and HIF1A, underscoring their potential as therapeutic agents for lung cancer. Conclusion: This study not only highlights the bioactive compounds of P. longum but also reinforces the molecular underpinnings of its anticancer mechanism, paving the way for future lung cancer therapeutics.
Collapse
Affiliation(s)
| | - Ashwath Kumar Balu
- Department of Biotechnology, PSG College of Technology, Peelamedu, Coimbatore, India
| | - Atul Shiju
- Department of Biotechnology, PSG College of Technology, Peelamedu, Coimbatore, India
| | - Pandiyan Muthuramalingam
- Division of Horticultural Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52725, Korea
| | - Hyunsuk Shin
- Division of Horticultural Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52725, Korea
| | - Baskar Venkidasamy
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, India
| | - Naiyf S. Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Shine Kadaikunnan
- Department of Botany and Microbiology, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Saunders A, Harrington PDB. Advances in Activity/Property Prediction from Chemical Structures. Crit Rev Anal Chem 2024; 54:135-147. [PMID: 35482792 DOI: 10.1080/10408347.2022.2066461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Recent technological advancement in AI modeling of molecular property databases has significantly expanded the opportunities for drug design and development. Quantitative structure-activity relationships (QSARs) are shown to provide more accurate predictions with regards to biological activity as well as toxicological assessment. By using a combination of in-silico models or by combining disparate structure-activity databases, researchers have been able to improve accuracy for a variety of drug discovery and analysis methods, generating viable compounds, which in certain cases, can be synthesized and further studied in vitro to find candidates for potential development. Additionally, the development of compounds of determined toxicology can be discontinued earlier, allowing alternative routes to be evaluated, preventing wasted time and resources. Although the progress that has been made is tremendous, expert review is still necessary for most in-silico generated predictions. Regardless, the scientific community continues to move ever closer to completely automated drug discovery and evaluation.
Collapse
Affiliation(s)
- Arianne Saunders
- Department of Chemistry and Biochemistry, Ohio University, Athens, Ohio, USA
| | | |
Collapse
|
8
|
Das AP, Agarwal SM. Recent advances in the area of plant-based anti-cancer drug discovery using computational approaches. Mol Divers 2024; 28:901-925. [PMID: 36670282 PMCID: PMC9859751 DOI: 10.1007/s11030-022-10590-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/18/2022] [Indexed: 01/22/2023]
Abstract
Phytocompounds are a well-established source of drug discovery due to their unique chemical and functional diversities. In the area of cancer therapeutics, several phytocompounds have been used till date to design and develop new drugs. One of the desired interests of pharmaceutical companies and researchers globally is that new anti-cancer leads are discovered, for which phytocompounds can be considered a valuable source. Simultaneously, in recent years, the growth of computational approaches like virtual screening (VS), molecular dynamics (MD), pharmacophore modelling, Quantitative structure-activity relationship (QSAR), Absorption Distribution Metabolism Excretion and Toxicity (ADMET), network biology, and machine learning (ML) has gained importance due to their efficiency, reduced time-consuming nature, and cost-effectiveness. Therefore, the present review amalgamates the information on plant-based molecules identified for cancer lead discovery from in silico approaches. The mandate of this review is to discuss studies published in the last 5-6 years that aim to identify the phytomolecules as leads against cancer with the help of traditional computational approaches as well as newer techniques like network pharmacology and ML. This review also lists the databases and webservers available in the public domain for phytocompounds related information that can be harnessed for drug discovery. It is expected that the present review would be useful to pharmacologists, medicinal chemists, molecular biologists, and other researchers involved in the development of natural products (NPs) into clinically effective lead molecules.
Collapse
Affiliation(s)
- Agneesh Pratim Das
- Bioinformatics Division, ICMR-National Institute of Cancer Prevention and Research, I-7, Sector-39, Noida, Uttar Pradesh, 201301, India
| | - Subhash Mohan Agarwal
- Bioinformatics Division, ICMR-National Institute of Cancer Prevention and Research, I-7, Sector-39, Noida, Uttar Pradesh, 201301, India.
| |
Collapse
|
9
|
Hassan J, Saeed SM, Deka L, Uddin MJ, Das DB. Applications of Machine Learning (ML) and Mathematical Modeling (MM) in Healthcare with Special Focus on Cancer Prognosis and Anticancer Therapy: Current Status and Challenges. Pharmaceutics 2024; 16:260. [PMID: 38399314 PMCID: PMC10892549 DOI: 10.3390/pharmaceutics16020260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
The use of data-driven high-throughput analytical techniques, which has given rise to computational oncology, is undisputed. The widespread use of machine learning (ML) and mathematical modeling (MM)-based techniques is widely acknowledged. These two approaches have fueled the advancement in cancer research and eventually led to the uptake of telemedicine in cancer care. For diagnostic, prognostic, and treatment purposes concerning different types of cancer research, vast databases of varied information with manifold dimensions are required, and indeed, all this information can only be managed by an automated system developed utilizing ML and MM. In addition, MM is being used to probe the relationship between the pharmacokinetics and pharmacodynamics (PK/PD interactions) of anti-cancer substances to improve cancer treatment, and also to refine the quality of existing treatment models by being incorporated at all steps of research and development related to cancer and in routine patient care. This review will serve as a consolidation of the advancement and benefits of ML and MM techniques with a special focus on the area of cancer prognosis and anticancer therapy, leading to the identification of challenges (data quantity, ethical consideration, and data privacy) which are yet to be fully addressed in current studies.
Collapse
Affiliation(s)
- Jasmin Hassan
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (S.M.S.)
| | | | - Lipika Deka
- Faculty of Computing, Engineering and Media, De Montfort University, Leicester LE1 9BH, UK;
| | - Md Jasim Uddin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Diganta B. Das
- Department of Chemical Engineering, Loughborough University, Loughborough LE11 3TU, UK
| |
Collapse
|
10
|
Arora P, Behera M, Saraf SA, Shukla R. Leveraging Artificial Intelligence for Synergies in Drug Discovery: From Computers to Clinics. Curr Pharm Des 2024; 30:2187-2205. [PMID: 38874046 DOI: 10.2174/0113816128308066240529121148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 06/15/2024]
Abstract
Over the period of the preceding decade, artificial intelligence (AI) has proved an outstanding performance in entire dimensions of science including pharmaceutical sciences. AI uses the concept of machine learning (ML), deep learning (DL), and neural networks (NNs) approaches for novel algorithm and hypothesis development by training the machines in multiple ways. AI-based drug development from molecule identification to clinical approval tremendously reduces the cost of development and the time over conventional methods. The COVID-19 vaccine development and approval by regulatory agencies within 1-2 years is the finest example of drug development. Hence, AI is fast becoming a boon for scientific researchers to streamline their advanced discoveries. AI-based FDA-approved nanomedicines perform well as target selective, synergistic therapies, recolonize the theragnostic pharmaceutical stream, and significantly improve drug research outcomes. This comprehensive review delves into the fundamental aspects of AI along with its applications in the realm of pharmaceutical life sciences. It explores AI's role in crucial areas such as drug designing, drug discovery and development, traditional Chinese medicine, integration of multi-omics data, as well as investigations into drug repurposing and polypharmacology studies.
Collapse
Affiliation(s)
- Priyanka Arora
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Near CRPF Base Camp, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow (UP)-226002, India
| | - Manaswini Behera
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Near CRPF Base Camp, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow (UP)-226002, India
| | - Shubhini A Saraf
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Near CRPF Base Camp, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow (UP)-226002, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Near CRPF Base Camp, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow (UP)-226002, India
| |
Collapse
|
11
|
Castillo-García EL, Cossio-Ramírez AL, Córdoba-Méndez ÓA, Loza-Mejía MA, Salazar JR, Chávez-Gutiérrez E, Bautista-Poblet G, Castillo-Mendieta NT, Moreno DA, García-Viguera C, Pinto-Almazán R, Almanza-Pérez JC, Gallardo JM, Guerra-Araiza C. In Silico and In Vivo Evaluation of the Maqui Berry ( Aristotelia chilensis (Mol.) Stuntz) on Biochemical Parameters and Oxidative Stress Markers in a Metabolic Syndrome Model. Metabolites 2023; 13:1189. [PMID: 38132871 PMCID: PMC10744843 DOI: 10.3390/metabo13121189] [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: 10/28/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023] Open
Abstract
Metabolic syndrome (MetS) is a complex disease that includes metabolic and physiological alterations in various organs such as the heart, pancreas, liver, and brain. Reports indicate that blackberry consumption, such as maqui berry, has a beneficial effect on chronic diseases such as cardiovascular disease, obesity, and diabetes. In the present study, in vivo and in silico studies have been performed to evaluate the molecular mechanisms implied to improve the metabolic parameters of MetS. Fourteen-day administration of maqui berry reduces weight gain, blood fasting glucose, total blood cholesterol, triacylglycerides, insulin resistance, and blood pressure impairment in the diet-induced MetS model in male and female rats. In addition, in the serum of male and female rats, the administration of maqui berry (MB) improved the concentration of MDA, the activity of SOD, and the formation of carbonyls in the group subjected to the diet-induced MetS model. In silico studies revealed that delphinidin and its glycosylated derivatives could be ligands of some metabolic targets such as α-glucosidase, PPAR-α, and PPAR-γ, which are related to MetS parameters. The experimental results obtained in the study suggest that even at low systemic concentrations, anthocyanin glycosides and aglycones could simultaneously act on different targets related to MetS. Therefore, these molecules could be used as coadjuvants in pharmacological interventions or as templates for designing new multitarget molecules to manage patients with MetS.
Collapse
Affiliation(s)
- Emily Leonela Castillo-García
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (E.L.C.-G.); (G.B.-P.)
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City 52919, Mexico
| | - Ana Lizzet Cossio-Ramírez
- Maestría en Ciencias de la Salud, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Óscar Arturo Córdoba-Méndez
- Design, Isolation, and Synthesis of Bioactive Molecules Research Group, Chemical Sciences School, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico; (Ó.A.C.-M.); (M.A.L.-M.); (J.R.S.)
| | - Marco A. Loza-Mejía
- Design, Isolation, and Synthesis of Bioactive Molecules Research Group, Chemical Sciences School, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico; (Ó.A.C.-M.); (M.A.L.-M.); (J.R.S.)
| | - Juan Rodrigo Salazar
- Design, Isolation, and Synthesis of Bioactive Molecules Research Group, Chemical Sciences School, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico; (Ó.A.C.-M.); (M.A.L.-M.); (J.R.S.)
| | - Edwin Chávez-Gutiérrez
- Doctorado en Ciencias en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación Manuel Carpio y Plan de Ayala s/n, Mexico City 11340, Mexico;
| | - Guadalupe Bautista-Poblet
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (E.L.C.-G.); (G.B.-P.)
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City 52919, Mexico
| | - Nadia Tzayaka Castillo-Mendieta
- Postdoctorate-Conacyt-Unidad de Investigación Médica en Enfermedades Neurologicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330 Col. Doctores, Mexico City 06725, Mexico;
| | - Diego A. Moreno
- Laboratorio de Fitoquímica y Alimentos Saludables (LabFAS), CEBAS, CSIC. Campus Universitario de Espinardo-25, E-30100 Murcia, Spain; (D.A.M.); (C.G.-V.)
| | - Cristina García-Viguera
- Laboratorio de Fitoquímica y Alimentos Saludables (LabFAS), CEBAS, CSIC. Campus Universitario de Espinardo-25, E-30100 Murcia, Spain; (D.A.M.); (C.G.-V.)
| | - Rodolfo Pinto-Almazán
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Mexico City 11340, Mexico
| | - Julio César Almanza-Pérez
- Laboratorio de Farmacologia, Departamento de Ciencias de la Salud, DCBS, UAM-I, Mexico City 09310, Mexico;
| | - Juan Manuel Gallardo
- Unidad de Investigación Médica en Enfermedades Nefrológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Christian Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (E.L.C.-G.); (G.B.-P.)
| |
Collapse
|
12
|
Parvatikar PP, Patil S, Khaparkhuntikar K, Patil S, Singh PK, Sahana R, Kulkarni RV, Raghu AV. Artificial intelligence: Machine learning approach for screening large database and drug discovery. Antiviral Res 2023; 220:105740. [PMID: 37935248 DOI: 10.1016/j.antiviral.2023.105740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/09/2023]
Abstract
Recent research in drug discovery dealing with many faces difficulties, including development of new drugs during disease outbreak and drug resistance due to rapidly accumulating mutations. Virtual screening is the most widely used method in computer aided drug discovery. It has a prominent ability in screening drug targets from large molecular databases. Recently, a number of web servers have developed for quickly screening publicly accessible chemical databases. In a nutshell, deep learning algorithms and artificial neural networks have modernised the field. Several drug discovery processes have used machine learning and deep learning algorithms, including peptide synthesis, structure-based virtual screening, ligand-based virtual screening, toxicity prediction, drug monitoring and release, pharmacophore modelling, quantitative structure-activity relationship, drug repositioning, polypharmacology, and physiochemical activity. Although there are presently a wide variety of data-driven AI/ML tools available, the majority of these tools have, up to this point, been developed in the context of non-communicable diseases like cancer, and a number of obstacles have prevented the translation of these tools to the discovery of treatments against infectious diseases. In this review various aspects of AI and ML in virtual screening of large databases were discussed. Here, with an emphasis on antivirals as well as other disease, offers a perspective on the advantages, drawbacks, and hazards of AI/ML techniques in the search for innovative treatments.
Collapse
Affiliation(s)
- Prachi P Parvatikar
- Department of Biotechnology, Allied Health Science, BLDE (Deemed-to-be University), Vijayapur 586103, Karnataka, India.
| | - Sudha Patil
- Department of Pharmaceutics, BLDEA's SSM College of Pharmacy and Research Centre, Vijayapur 586 103, Karnataka, India
| | - Kedar Khaparkhuntikar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Shruti Patil
- Department of Biotechnology, Allied Health Science, BLDE (Deemed-to-be University), Vijayapur 586103, Karnataka, India
| | - Pankaj K Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - R Sahana
- Department of Computer Science and Engineering, RV Institute of Technology and Management, 560076, Bengaluru, India
| | - Raghavendra V Kulkarni
- Department of Biotechnology, Allied Health Science, BLDE (Deemed-to-be University), Vijayapur 586103, Karnataka, India; Department of Pharmaceutics, BLDEA's SSM College of Pharmacy and Research Centre, Vijayapur 586 103, Karnataka, India
| | - Anjanapura V Raghu
- Department of Science and Technology, BLDE (Deemed-to-be University), Vijayapur 586103, Karnataka, India.
| |
Collapse
|
13
|
Cui Z, Zhang Y, Zhang Z, Abudurexiti A, Yusuf A. Synthesis of an aggregation-induced emission-based fluorescent probe based on rupestonic acid. RSC Adv 2023; 13:25369-25378. [PMID: 37661955 PMCID: PMC10472508 DOI: 10.1039/d3ra03521b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/14/2023] [Indexed: 09/05/2023] Open
Abstract
Chinese herbal medicine and Chinese patent medicine have been widely applied for cancer care in China. Rupestonic acid, an active ingredient of Artemisia rupestris L., has recently been confirmed to have certain anti-tumor effects in vitro. In this study, we employed the application of a commonly devoted triphenylamine as a fluorophore and the addition of 2,4-thiazolidinedione as a bridge to integrate rupestonic acid into the AIE system to create an fluorescent probe with anti-tumor properties. The spectral, cytotoxic, and cellular imaging properties of the probe were measured. Its promising responses make possible the application of the probe in antitumor theragnostic systems.
Collapse
Affiliation(s)
- Zhichao Cui
- College of Chemistry and Environmental Science, Laboratory of Xinjiang Native Medicinal and Edible Plant Resources Chemistry, Kashi University Kashi 844000 China +86-18690293325
| | - Yucai Zhang
- College of Chemistry and Environmental Science, Laboratory of Xinjiang Native Medicinal and Edible Plant Resources Chemistry, Kashi University Kashi 844000 China +86-18690293325
| | - Zhonghui Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University Guangzhou 510006 China
| | - Adila Abudurexiti
- College of Chemistry and Environmental Science, Laboratory of Xinjiang Native Medicinal and Edible Plant Resources Chemistry, Kashi University Kashi 844000 China +86-18690293325
| | - Abdulla Yusuf
- College of Chemistry and Environmental Science, Laboratory of Xinjiang Native Medicinal and Edible Plant Resources Chemistry, Kashi University Kashi 844000 China +86-18690293325
| |
Collapse
|
14
|
Cunningham M, Pins D, Dezső Z, Torrent M, Vasanthakumar A, Pandey A. PINNED: identifying characteristics of druggable human proteins using an interpretable neural network. J Cheminform 2023; 15:64. [PMID: 37468968 PMCID: PMC10354961 DOI: 10.1186/s13321-023-00735-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
The identification of human proteins that are amenable to pharmacologic modulation without significant off-target effects remains an important unsolved challenge. Computational methods have been devised to identify features which distinguish between "druggable" and "undruggable" proteins, finding that protein sequence, tissue and cellular localization, biological role, and position in the protein-protein interaction network are all important discriminant factors. However, many prior efforts to automate the assessment of protein druggability suffer from low performance or poor interpretability. We developed a neural network-based machine learning model capable of generating druggability sub-scores based on each of four distinct categories, combining them to form an overall druggability score. The model achieves an excellent performance in separating drugged and undrugged proteins in the human proteome, with an area under the receiver operating characteristic (AUC) of 0.95. Our use of multiple sub-scores allows the assessment of potential protein targets of interest based on distinct contributors to druggability, leading to a more interpretable and holistic model to identify novel targets.
Collapse
Affiliation(s)
- Michael Cunningham
- Genomics Research Center, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL, 60064, USA.
| | - Danielle Pins
- Information Research, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL, 60064, USA
| | - Zoltán Dezső
- Genomics Research Center, AbbVie Inc., 1000 Gateway Boulevard, South San Francisco, CA, 94080, USA
| | - Maricel Torrent
- Small Molecule Therapeutics and Platform Technologies, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL, 60064, USA
| | - Aparna Vasanthakumar
- Genomics Research Center, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL, 60064, USA
| | - Abhishek Pandey
- Information Research, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL, 60064, USA
| |
Collapse
|
15
|
Narganes-Carlón D, Crowther DJ, Pearson ER. A publication-wide association study (PWAS), historical language models to prioritise novel therapeutic drug targets. Sci Rep 2023; 13:8366. [PMID: 37225853 DOI: 10.1038/s41598-023-35597-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/20/2023] [Indexed: 05/26/2023] Open
Abstract
Most biomedical knowledge is published as text, making it challenging to analyse using traditional statistical methods. In contrast, machine-interpretable data primarily comes from structured property databases, which represent only a fraction of the knowledge present in the biomedical literature. Crucial insights and inferences can be drawn from these publications by the scientific community. We trained language models on literature from different time periods to evaluate their ranking of prospective gene-disease associations and protein-protein interactions. Using 28 distinct historical text corpora of abstracts published between 1995 and 2022, we trained independent Word2Vec models to prioritise associations that were likely to be reported in future years. This study demonstrates that biomedical knowledge can be encoded as word embeddings without the need for human labelling or supervision. Language models effectively capture drug discovery concepts such as clinical tractability, disease associations, and biochemical pathways. Additionally, these models can prioritise hypotheses years before their initial reporting. Our findings underscore the potential for extracting yet-to-be-discovered relationships through data-driven approaches, leading to generalised biomedical literature mining for potential therapeutic drug targets. The Publication-Wide Association Study (PWAS) enables the prioritisation of under-explored targets and provides a scalable system for accelerating early-stage target ranking, irrespective of the specific disease of interest.
Collapse
Affiliation(s)
- David Narganes-Carlón
- Division of Population Health and Genomics, Ninewells Hospital, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
- Exscientia Ltd, Dundee One, River Court, 5 West Victoria Dock Road, Dundee, DD1 3JT, UK.
| | - Daniel J Crowther
- Exscientia Ltd, Dundee One, River Court, 5 West Victoria Dock Road, Dundee, DD1 3JT, UK
| | - Ewan R Pearson
- Division of Population Health and Genomics, Ninewells Hospital, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| |
Collapse
|
16
|
Carneiro J, Magalhães RP, de la Oliva Roque VM, Simões M, Pratas D, Sousa SF. TargIDe: a machine-learning workflow for target identification of molecules with antibiofilm activity against Pseudomonas aeruginosa. J Comput Aided Mol Des 2023; 37:265-278. [PMID: 37085636 DOI: 10.1007/s10822-023-00505-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/12/2023] [Indexed: 04/23/2023]
Abstract
Bacterial biofilms are a source of infectious human diseases and are heavily linked to antibiotic resistance. Pseudomonas aeruginosa is a multidrug-resistant bacterium widely present and implicated in several hospital-acquired infections. Over the last years, the development of new drugs able to inhibit Pseudomonas aeruginosa by interfering with its ability to form biofilms has become a promising strategy in drug discovery. Identifying molecules able to interfere with biofilm formation is difficult, but further developing these molecules by rationally improving their activity is particularly challenging, as it requires knowledge of the specific protein target that is inhibited. This work describes the development of a machine learning multitechnique consensus workflow to predict the protein targets of molecules with confirmed inhibitory activity against biofilm formation by Pseudomonas aeruginosa. It uses a specialized database containing all the known targets implicated in biofilm formation by Pseudomonas aeruginosa. The experimentally confirmed inhibitors available on ChEMBL, together with chemical descriptors, were used as the input features for a combination of nine different classification models, yielding a consensus method to predict the most likely target of a ligand. The implemented algorithm is freely available at https://github.com/BioSIM-Research-Group/TargIDe under licence GNU General Public Licence (GPL) version 3 and can easily be improved as more data become available.
Collapse
Affiliation(s)
- João Carneiro
- Interdisciplinary Centre of Marine and Environmental Research, CIIMAR, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, Porto, 4450-208, Portugal.
| | - Rita P Magalhães
- Faculty of Medicine, Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4200-319, Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, UCIBIO-Applied Molecular Biosciences Unit, University of Porto, BioSIM, Porto, 4200-319, Portugal
| | - Victor M de la Oliva Roque
- Faculty of Medicine, Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4200-319, Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, UCIBIO-Applied Molecular Biosciences Unit, University of Porto, BioSIM, Porto, 4200-319, Portugal
| | - Manuel Simões
- Faculty of Engineering, LEPABE Laboratory for Process Engineering, Environment, Biotechnology and Energy, University of Porto, Rua Dr. Roberto Frias, s/n, Porto, 4200-465, Portugal
- Faculty of Engineering, ALiCE-Associate Laboratory in Chemical Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
| | - Diogo Pratas
- Institute of Electronics and Informatics Engineering of Aveiro, IEETA, University of Aveiro, Aveiro, Portugal
- Department of Electronics, Telecommunications and Informatics, DETI, University of Aveiro, Aveiro, Portugal
- Department of Virology, DoV, University of Helsinki, Helsinki, Finland
| | - Sérgio F Sousa
- Faculty of Medicine, Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4200-319, Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, UCIBIO-Applied Molecular Biosciences Unit, University of Porto, BioSIM, Porto, 4200-319, Portugal
| |
Collapse
|
17
|
Cavasotto CN, Di Filippo JI. The Impact of Supervised Learning Methods in Ultralarge High-Throughput Docking. J Chem Inf Model 2023; 63:2267-2280. [PMID: 37036491 DOI: 10.1021/acs.jcim.2c01471] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Structure-based virtual screening methods are, nowadays, one of the key pillars of computational drug discovery. In recent years, a series of studies have reported docking-based virtual screening campaigns of large databases ranging from hundreds to thousands of millions compounds, further identifying novel hits after experimental validation. As these larg-scale efforts are not generally accessible, machine learning-based protocols have emerged to accelerate the identification of virtual hits within an ultralarge chemical space, reaching impressive reductions in computational time. Herein, we illustrate the motivation and the problem behind the screening of large databases, providing an overview of key concepts and essential applications of machine learning-accelerated protocols, specifically concerning supervised learning methods. We also discuss where the field stands with these novel developments, highlighting possible insights for future studies.
Collapse
Affiliation(s)
- Claudio N Cavasotto
- Computational Drug Design and Biomedical Informatics Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Juan Domingo Perón 1500, B1629AHJ Pilar, Argentina
- Facultad de Ciencias Biomédicas, and Facultad de Ingeniería, Universidad Austral, Av. Juan Domingo Perón 1500, B1629AHJ Pilar, Argentina
- Austral Institute for Applied Artificial Intelligence, Universidad Austral, Av. Juan Domingo Perón 1500, B1629AHJ Pilar, Argentina
| | - Juan I Di Filippo
- Computational Drug Design and Biomedical Informatics Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Juan Domingo Perón 1500, B1629AHJ Pilar, Argentina
- Facultad de Ciencias Biomédicas, and Facultad de Ingeniería, Universidad Austral, Av. Juan Domingo Perón 1500, B1629AHJ Pilar, Argentina
- Austral Institute for Applied Artificial Intelligence, Universidad Austral, Av. Juan Domingo Perón 1500, B1629AHJ Pilar, Argentina
| |
Collapse
|
18
|
Thafar MA, Albaradei S, Uludag M, Alshahrani M, Gojobori T, Essack M, Gao X. OncoRTT: Predicting novel oncology-related therapeutic targets using BERT embeddings and omics features. Front Genet 2023; 14:1139626. [PMID: 37091791 PMCID: PMC10117673 DOI: 10.3389/fgene.2023.1139626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
Late-stage drug development failures are usually a consequence of ineffective targets. Thus, proper target identification is needed, which may be possible using computational approaches. The reason being, effective targets have disease-relevant biological functions, and omics data unveil the proteins involved in these functions. Also, properties that favor the existence of binding between drug and target are deducible from the protein’s amino acid sequence. In this work, we developed OncoRTT, a deep learning (DL)-based method for predicting novel therapeutic targets. OncoRTT is designed to reduce suboptimal target selection by identifying novel targets based on features of known effective targets using DL approaches. First, we created the “OncologyTT” datasets, which include genes/proteins associated with ten prevalent cancer types. Then, we generated three sets of features for all genes: omics features, the proteins’ amino-acid sequence BERT embeddings, and the integrated features to train and test the DL classifiers separately. The models achieved high prediction performances in terms of area under the curve (AUC), i.e., AUC greater than 0.88 for all cancer types, with a maximum of 0.95 for leukemia. Also, OncoRTT outperformed the state-of-the-art method using their data in five out of seven cancer types commonly assessed by both methods. Furthermore, OncoRTT predicts novel therapeutic targets using new test data related to the seven cancer types. We further corroborated these results with other validation evidence using the Open Targets Platform and a case study focused on the top-10 predicted therapeutic targets for lung cancer.
Collapse
Affiliation(s)
- Maha A. Thafar
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center, Computer (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- College of Computers and Information Technology, Computer Science Department, Taif University, Taif, Saudi Arabia
| | - Somayah Albaradei
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center, Computer (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Faculty of Computing and Information Technology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmut Uludag
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center, Computer (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Mona Alshahrani
- National Center for Artificial Intelligence (NCAI), Saudi Data and Artificial Intelligence Authority (SDAIA), Riyadh, Saudi Arabia
| | - Takashi Gojobori
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center, Computer (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Magbubah Essack
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center, Computer (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- *Correspondence: Xin Gao, ; Magbubah Essack,
| | - Xin Gao
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center, Computer (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- *Correspondence: Xin Gao, ; Magbubah Essack,
| |
Collapse
|
19
|
Voitalov I, Zhang L, Kilpatrick C, Withers JB, Saleh A, Akmaev VR, Ghiassian SD. The module triad: a novel network biology approach to utilize patients' multi-omics data for target discovery in ulcerative colitis. Sci Rep 2022; 12:21685. [PMID: 36522454 PMCID: PMC9755270 DOI: 10.1038/s41598-022-26276-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Tumor necrosis factor-[Formula: see text] inhibitors (TNFi) have been a standard treatment in ulcerative colitis (UC) for nearly 20 years. However, insufficient response rate to TNFi therapies along with concerns around their immunogenicity and inconvenience of drug delivery through injections calls for development of UC drugs targeting alternative proteins. Here, we propose a multi-omic network biology method for prioritization of protein targets for UC treatment. Our method identifies network modules on the Human Interactome-a network of protein-protein interactions in human cells-consisting of genes contributing to the predisposition to UC (Genotype module), genes whose expression needs to be modulated to achieve low disease activity (Response module), and proteins whose perturbation alters expression of the Response module genes to a healthy state (Treatment module). Targets are prioritized based on their topological relevance to the Genotype module and functional similarity to the Treatment module. We demonstrate utility of our method in UC and other complex diseases by efficiently recovering the protein targets associated with compounds in clinical trials and on the market . The proposed method may help to reduce cost and time of drug development by offering a computational screening tool for identification of novel and repurposing therapeutic opportunities in UC and other complex diseases.
Collapse
Affiliation(s)
- Ivan Voitalov
- Scipher Medicine Corporation, 221 Crescent St Suite 103A, Waltham, MA 02453 USA
| | - Lixia Zhang
- Scipher Medicine Corporation, 221 Crescent St Suite 103A, Waltham, MA 02453 USA
| | - Casey Kilpatrick
- Scipher Medicine Corporation, 221 Crescent St Suite 103A, Waltham, MA 02453 USA
| | - Johanna B. Withers
- Scipher Medicine Corporation, 221 Crescent St Suite 103A, Waltham, MA 02453 USA
| | - Alif Saleh
- Scipher Medicine Corporation, 221 Crescent St Suite 103A, Waltham, MA 02453 USA
| | | | | |
Collapse
|
20
|
Raza MA, Aziz S, Noreen M, Saeed A, Anjum I, Ahmed M, Raza SM. Artificial Intelligence (AI) in Pharmacy: An Overview of Innovations. Innov Pharm 2022; 13:10.24926/iip.v13i2.4839. [PMID: 36654703 PMCID: PMC9836757 DOI: 10.24926/iip.v13i2.4839] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Artificial Intelligence (AI) emerged as an intervention for data and number-related problems. This breakthrough has led to several technological advancements in virtually all fields from engineering to architecture, education, accounting, business, health, and so on. AI has come a long way in healthcare, having played significant roles in data and information storage and management - such as patient medical histories, medicine stocks, sale records, and so on; automated machines; software and computer applications like diagnostic tools such as MRI radiation technology, CT diagnosis and many more have all been created to aid and simplify healthcare measures. Inarguably, AI has revolutionized healthcare to be more effective and efficient and the pharmacy sector is not left out. During the past few years, a considerable amount of increasing interest in the uses of AI technology has been identified for analyzing as well as interpreting some important fields of pharmacy like drug discovery, dosage form designing, polypharmacology, and hospital pharmacy. Given the growing importance of AI, we wanted to create a comprehensive report which helps every practicing pharmacist understand the biggest breakthroughs which are assisted by the deployment of this field.
Collapse
Affiliation(s)
- Muhammad Ahmer Raza
- Department of Pharmacy Practice, The University of Lahore, Punjab, Pakistan,Faculty of Pharmacy, The University of Faisalabad, Punjab, Pakistan
| | - Shireen Aziz
- School of Pharmacy, Zhengzhou University, Henan, China,Faculty of Pharmacy, University of Sargodha, Punjab, Pakistan
| | - Misbah Noreen
- Faculty of Pharmacy, The University of Faisalabad, Punjab, Pakistan,School of Pharmacy, University of Agriculture, Faisalabad, Punjab, Pakistan
| | - Amna Saeed
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmacy, Xi’an Jiaotong University, Xi’an, China,Center for Drug Safety and Policy Research, Xi’an Jiaotong University, Xi’an, China
| | - Irfan Anjum
- Faculty of Pharmacy, The University of Lahore, Pakistan,Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Mudassar Ahmed
- Faculty of Pharmacy, The University of Faisalabad, Punjab, Pakistan,School of Pharmacy, University of Agriculture, Faisalabad, Punjab, Pakistan
| | - Shahid Masood Raza
- Faculty of Pharmacy, The University of Faisalabad, Punjab, Pakistan,Faculty of Pharmacy, University of Sargodha, Punjab, Pakistan,School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China,Corresponding author: Shahid Masood Raza School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| |
Collapse
|
21
|
Mostofa F, Yasid NA, Shamsi S, Ahmad SA, Mohd-Yusoff NF, Abas F, Ahmad S. In Silico Study and Effects of BDMC33 on TNBS-Induced BMP Gene Expressions in Zebrafish Gut Inflammation-Associated Arthritis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238304. [PMID: 36500396 PMCID: PMC9740523 DOI: 10.3390/molecules27238304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022]
Abstract
The bone morphogenic protein (BMP) family is a member of the TGF-beta superfamily and plays a crucial role during the onset of gut inflammation and arthritis diseases. Recent studies have reported a connection with the gut-joint axis; however, the genetic players are still less explored. Meanwhile, BDMC33 is a newly synthesized anti-inflammatory drug candidate. Therefore, in our present study, we analysed the genome-wide features of the BMP family as well as the role of BMP members in gut-associated arthritis in an inflammatory state and the ability of BDMC33 to attenuate this inflammation. Firstly, genome-wide analyses were performed on the BMP family in the zebrafish genome, employing several in silico techniques. Afterwards, the effects of curcumin analogues on BMP gene expression in zebrafish larvae induced with TNBS (0.78 mg/mL) were determined using real time-qPCR. A total of 38 identified BMP proteins were revealed to be clustered in five major clades and contain TGF beta and TGF beta pro peptide domains. Furthermore, BDMC33 suppressed the expression of four selected BMP genes in the TNBS-induced larvae, where the highest gene suppression was in the BMP2a gene (an eight-fold decrement), followed by BMP7b (four-fold decrement), BMP4 (four-fold decrement), and BMP6 (three-fold decrement). Therefore, this study reveals the role of BMPs in gut-associated arthritis and proves the ability of BDMC33 to act as a potential anti-inflammatory drug for suppressing TNBS-induced BMP genes in zebrafish larvae.
Collapse
Affiliation(s)
- Farhana Mostofa
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Nur Adeela Yasid
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Suhaili Shamsi
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Siti Aqlima Ahmad
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Nur Fatihah Mohd-Yusoff
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Bimolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Faridah Abas
- Department of Food Science, Faculty of Food Science & Technology, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Syahida Ahmad
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Correspondence: ; Tel.: +603-97696724
| |
Collapse
|
22
|
Mohammed AE, Alghamdi SS, Alharbi NK, Alshehri F, Suliman RS, Al-Dhabaan F, Alharbi M. Limoniastrum monopetalum-Mediated Nanoparticles and Biomedicines: In Silico Study and Molecular Prediction of Biomolecules. Molecules 2022; 27:8014. [PMID: 36432115 PMCID: PMC9693928 DOI: 10.3390/molecules27228014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/05/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022] Open
Abstract
An in silico approach applying computer-simulated models helps enhance biomedicines by sightseeing the pharmacology of potential therapeutics. Currently, an in silico study combined with in vitro assays investigated the antimicrobial ability of Limoniastrum monopetalum and silver nanoparticles (AgNPs) fabricated by its aid. AgNPs mediated by L. monopetalum were characterized using FTIR, TEM, SEM, and DLS. L. monopetalum metabolites were detected by QTOF-LCMS and assessed using an in silico study for pharmacological properties. The antibacterial ability of an L. monopetalum extract and AgNPs was investigated. PASS Online predictions and the swissADME web server were used for antibacterial activity and potential molecular target metabolites, respectively. Spherical AgNPs with a 68.79 nm average size diameter were obtained. Twelve biomolecules (ferulic acid, trihydroxy-octadecenoic acid, catechin, pinoresinol, gallic acid, myricetin, 6-hydroxyluteolin, 6,7-dihydroxy-5-methoxy 7-O-β-d-glucopyranoside, methyl gallate, isorhamnetin, chlorogenic acid, 2-(3,4-dihydroxyphenyl)-5,7-dihydroxy-4-oxo-4H-chromen-3-yl 6-O-(6-deoxy-β-l-mannopyranosyl)-β-d-glucopyranoside) were identified. The L. monopetalum extract and AgNPs displayed antibacterial effects. The computational study suggested that L. Monopetalum metabolites could hold promising antibacterial activity with minimal toxicity and an acceptable pharmaceutical profile. The in silico approach indicated that metabolites 8 and 12 have the highest antibacterial activity, and swissADME web server results suggested the CA II enzyme as a potential molecular target for both metabolites. Novel therapeutic agents could be discovered using in silico molecular target prediction combined with in vitro studies. Among L. Monopetalum metabolites, metabolite 12 could serve as a starting point for potential antibacterial treatment for several human bacterial infections.
Collapse
Affiliation(s)
- Afrah E. Mohammed
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Sahar S. Alghamdi
- Department of Pharmaceutical Sciences, College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Riyadh 11481, Saudi Arabia
| | - Nada K. Alharbi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Fatma Alshehri
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Rasha Saad Suliman
- Department of Pharmacy, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates
| | - Fahad Al-Dhabaan
- Department of Biology, College of Science and Humanities, Shaqra University, Ad-Dawadimi 11911, Saudi Arabia
| | - Maha Alharbi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| |
Collapse
|
23
|
Masoudi-Sobhanzadeh Y, Gholaminejad A, Gheisari Y, Roointan A. Discovering driver nodes in chronic kidney disease-related networks using Trader as a newly developed algorithm. Comput Biol Med 2022; 148:105892. [PMID: 35932730 DOI: 10.1016/j.compbiomed.2022.105892] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/04/2022] [Accepted: 07/16/2022] [Indexed: 11/18/2022]
Abstract
Thanks to the advances in the field of computational-based biology, a huge volume of disease-related data has been generated so far. From the existing data, the disease-related protein-protein interaction (PPI) networks seem to yield effective treatment plans due to the informative/systematic representation of diseases. Yet, a large number of previous studies have failed due to the complex nature of such disease-related networks. For addressing this limitation, in the present study, we combined Trader and the DFS algorithms to identify a minimal subset of nodes (driver nodes) whose removal produces a maximum number of disjoint sub-networks. We then screened the nodes in the disease-associated PPI networks and to evaluate the efficiency of the suggested method, it was applied to six PPI networks of differentially expressed genes in chronic kidney diseases. The performance of Trader was superior to other well-known algorithms in terms of identifying driver nodes. Besides, the proportion of proteins that were targeted by at least one FDA-approved drug was significantly higher among the identified driver nodes when compared with the rest of the proteins in the networks. The proposed algorithm could be applied for predicting future therapeutic targets in complex disorder networks. In conclusion, unlike the common methods, computationally efficient algorithms can generate more practical outcomes which are compatible with real-world biological facts.
Collapse
Affiliation(s)
- Yosef Masoudi-Sobhanzadeh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alieh Gholaminejad
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yousof Gheisari
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Roointan
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
24
|
Liu Z, Li H, Jin Z, Li Y, Guo F, He Y, Liu X, Qi Y, Yuan L, He F, Li D. Exploration of Target Spaces in the Human Genome for Protein and Peptide Drugs. GENOMICS, PROTEOMICS & BIOINFORMATICS 2022; 20:780-794. [PMID: 35338014 PMCID: PMC9881050 DOI: 10.1016/j.gpb.2021.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/20/2021] [Accepted: 11/01/2021] [Indexed: 01/31/2023]
Abstract
After decades of development, protein and peptide drugs have now grown into a major drug class in the marketplace. Target identification and validation are crucial for the discovery of protein and peptide drugs, and bioinformatics prediction of targets based on the characteristics of known target proteins will help improve the efficiency and success rate of target selection. However, owing to the developmental history in the pharmaceutical industry, previous systematic exploration of the target spaces has mainly focused on traditional small-molecule drugs, while studies related to protein and peptide drugs are lacking. Here, we systematically explore the target spaces in the human genome specifically for protein and peptide drugs. Compared with other proteins, both successful protein and peptide drug targets have many special characteristics, and are also significantly different from those of small-molecule drugs in many aspects. Based on these features, we develop separate effective genome-wide target prediction models for protein and peptide drugs. Finally, a user-friendly web server, Predictor Of Protein and PeptIde drugs' therapeutic Targets (POPPIT) (http://poppit.ncpsb.org.cn/), is established, which provides not only target prediction specifically for protein and peptide drugs but also abundant annotations for predicted targets.
Collapse
Affiliation(s)
- Zhongyang Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China,School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China,College of Chemistry and Environmental Science, Hebei University, Baoding 071002, China,Corresponding authors.
| | - Honglei Li
- Suzhou Geneworks Technology Co., Ltd., Suzhou 215028, China
| | - Zhaoyu Jin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yang Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Feifei Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yangzhige He
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Xinyue Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yaning Qi
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China,College of Life Sciences, Hebei University, Baoding 071002, China
| | - Liying Yuan
- College of Life Sciences, Hebei University, Baoding 071002, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China,Corresponding authors.
| | - Dong Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China,School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China,Corresponding authors.
| |
Collapse
|
25
|
Han Y, Klinger K, Rajpal DK, Zhu C, Teeple E. Empowering the discovery of novel target-disease associations via machine learning approaches in the open targets platform. BMC Bioinformatics 2022; 23:232. [PMID: 35710324 PMCID: PMC9202116 DOI: 10.1186/s12859-022-04753-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/26/2022] [Indexed: 11/10/2022] Open
Abstract
Background The Open Targets (OT) Platform integrates a wide range of data sources on target-disease associations to facilitate identification of potential therapeutic drug targets to treat human diseases. However, due to the complexity that targets are usually functionally pleiotropic and efficacious for multiple indications, challenges in identifying novel target to indication associations remain. Specifically, persistent need exists for new methods for integration of novel target-disease association evidence and biological knowledge bases via advanced computational methods. These offer promise for increasing power for identification of the most promising target-disease pairs for therapeutic development. Here we introduce a novel approach by integrating additional target-disease features with machine learning models to further uncover druggable disease to target indications. Results We derived novel target-disease associations as supplemental features to OT platform-based associations using three data sources: (1) target tissue specificity from GTEx expression profiles; (2) target semantic similarities based on gene ontology; and (3) functional interactions among targets by embedding them from protein–protein interaction (PPI) networks. Machine learning models were applied to evaluate feature importance and performance benchmarks for predicting targets with known drug indications. The evaluation results show the newly integrated features demonstrate higher importance than current features in OT. In addition, these also show superior performance over association benchmarks and may support discovery of novel therapeutic indications for highly pursued targets. Conclusion Our newly generated features can be used to represent additional underlying biological relatedness among targets and diseases to further empower improved performance for predicting novel indications for drug targets through advanced machine learning models. The proposed methodology enables a powerful new approach for systematic evaluation of drug targets with novel indications. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04753-4.
Collapse
Affiliation(s)
- Yingnan Han
- Translational Sciences, Sanofi US, Framingham, MA, 01701, USA
| | | | - Deepak K Rajpal
- Translational Sciences, Sanofi US, Framingham, MA, 01701, USA
| | - Cheng Zhu
- Translational Sciences, Sanofi US, Framingham, MA, 01701, USA.
| | - Erin Teeple
- Translational Sciences, Sanofi US, Framingham, MA, 01701, USA.
| |
Collapse
|
26
|
In-silico predicting as a tool to develop plant-based biomedicines and nanoparticles: Lycium shawii metabolites. Biomed Pharmacother 2022; 150:113008. [PMID: 35489282 DOI: 10.1016/j.biopha.2022.113008] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION AND PURPOSE In silico approach helps develop biomedicines and is useful for exploring the pharmacology of potential therapeutics using computer-simulated models. In vitro assays were used to determine the anti-microbial and cytotoxic efficacies of silver nanoparticles (AgNPs) synthesized with the shrub Lycium shawii. METHODS In silico predicting was performed to assess the L. shawii metabolites identified using QTOF-LCMS for their pharmacological properties. L. shawii mediated AgNPs were synthesized and characterized (FTIR, TEM, SEM, DLS and EDX). The anti-bacterial efficacies of L. shawii extract, AgNPs, and penicillin-conjugated AgNPs (pen-AgNPs) were determined. The cytotoxicity of the AgNPs was measured against colorectal cancer cell line (HCT116), normal breast epithelium (MCF 10 A), and breast cancer cell line (MDA MB 231). RESULTS AND DISCUSSION Five molecules (costunolide, catechin, emodin, lyciumaside, and aloe emodin 11-O-rhamnoside) were detected in the L. shawii extract. AgNPs (69 nm) were spherical with crystallographic structure. All three agents prepared showed inhibitory activity against the tested bacteria, the most efficacious being pen-AgNPs. High cytotoxicity of AgNPs (IC50 62 μg/ml) was observed against HCT116, IC50 was 78 μg/ml for MCF 10 A, and 250 μg/ml for MDA MB 231, of which cells showed apoptotic features under TEM examination. The in silico approach indicated that the carbonic anhydrase IX enzyme was the target molecule mediating anti-cancer and anti-bacterial activities and that emodin was the metabolite in action. CONCLUSIONS Combining in vitro studies and in silico molecular target prediction helps find novel therapeutic agents. Among L. shawii metabolites, emodin is suggested for further studies as an agent for drug development against pathogenic bacteria and cancer.
Collapse
|
27
|
In silico Methods for Identification of Potential Therapeutic Targets. Interdiscip Sci 2022; 14:285-310. [PMID: 34826045 PMCID: PMC8616973 DOI: 10.1007/s12539-021-00491-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 10/19/2021] [Accepted: 11/01/2021] [Indexed: 11/01/2022]
Abstract
AbstractAt the initial stage of drug discovery, identifying novel targets with maximal efficacy and minimal side effects can improve the success rate and portfolio value of drug discovery projects while simultaneously reducing cycle time and cost. However, harnessing the full potential of big data to narrow the range of plausible targets through existing computational methods remains a key issue in this field. This paper reviews two categories of in silico methods—comparative genomics and network-based methods—for finding potential therapeutic targets among cellular functions based on understanding their related biological processes. In addition to describing the principles, databases, software, and applications, we discuss some recent studies and prospects of the methods. While comparative genomics is mostly applied to infectious diseases, network-based methods can be applied to infectious and non-infectious diseases. Nonetheless, the methods often complement each other in their advantages and disadvantages. The information reported here guides toward improving the application of big data-driven computational methods for therapeutic target discovery.
Graphical abstract
Collapse
|
28
|
Khan A, Alsahli MA, Aljasir MA, Maswadeh H, Mobark MA, Azam F, Allemailem KS, Alrumaihi F, Alhumaydhi FA, Alwashmi ASS, Almatroudi AA, Alsugoor MH, Khan MA. Safety, Stability, and Therapeutic Efficacy of Long-Circulating TQ-Incorporated Liposomes: Implication in the Treatment of Lung Cancer. Pharmaceutics 2022; 14:153. [PMID: 35057049 PMCID: PMC8778344 DOI: 10.3390/pharmaceutics14010153] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/01/2022] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
Thymoquinone (TQ), which is one of the main bioactive constituents of Nigella sativa seeds, has demonstrated its potential against various cancer models. The poor solubility of TQ in aqueous solution limits its uses in clinical application. The present study aimed to develop a novel formulation of TQ to increase its bioavailability and therapeutic potential with minimal toxicity. Polyethylene glycol (PEG)-coated DSPC/cholesterol comprising TQ liposomes (PEG-Lip-TQ) were prepared and characterized on various aspects. A computational investigation using molecular docking was used to assess the possible binding interactions of TQ with 12 prospective anticancer drug targets. The in vitro anticancer activity was assessed in A549 and H460 lung cancer cells in a time- and dose-dependent manner, while the oral acute toxicity assay was evaluated in silico as well as in vivo in mice. TQ docked to the Hsp90 target had the lowest binding energy of -6.05 kcal/mol, whereas caspase 3 was recognized as the least likely target for TQ with a binding energy of -1.19 kcal/mol. The results showed 96% EE with 120 nm size, and -10.85 mv, ζ-potential of PEG-Lip-TQ, respectively. The cell cytotoxicity data demonstrated high sensitivity of PEG-Lip-TQ and a several fold decrease in the IC50 while comparing free TQ. The cell cycle analysis showed changes in the distribution of cells with doses. The in vivo data revealed an ~9-fold increase in the LD50 of PEG-Lip-TQ on free TQ as an estimated 775 and 89.5 mg/kg b.w, respectively. This study indicates that the pharmacological and efficacy profile of PEG-lip-TQ is superior to free TQ, which will pave the way for an exploration of the effect of TQ formulation in the treatment of lung cancer in clinical settings.
Collapse
Affiliation(s)
- Arif Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Mohammed A. Alsahli
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (M.A.A.); (M.A.A.); (K.S.A.); (F.A.); (F.A.A.); (A.S.S.A.); (A.A.A.)
| | - Mohammad A. Aljasir
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (M.A.A.); (M.A.A.); (K.S.A.); (F.A.); (F.A.A.); (A.S.S.A.); (A.A.A.)
| | - Hamzah Maswadeh
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Mugahid A. Mobark
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia;
- Department of Pathology, Faculty of Medicine, University of Kordofan, El-Obeid 157, Sudan
| | - Faizul Azam
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Unaizah 51911, Saudi Arabia;
| | - Khaled S. Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (M.A.A.); (M.A.A.); (K.S.A.); (F.A.); (F.A.A.); (A.S.S.A.); (A.A.A.)
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (M.A.A.); (M.A.A.); (K.S.A.); (F.A.); (F.A.A.); (A.S.S.A.); (A.A.A.)
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (M.A.A.); (M.A.A.); (K.S.A.); (F.A.); (F.A.A.); (A.S.S.A.); (A.A.A.)
| | - Ameen S. S. Alwashmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (M.A.A.); (M.A.A.); (K.S.A.); (F.A.); (F.A.A.); (A.S.S.A.); (A.A.A.)
| | - Ahmed A. Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (M.A.A.); (M.A.A.); (K.S.A.); (F.A.); (F.A.A.); (A.S.S.A.); (A.A.A.)
| | - Mahdi H. Alsugoor
- Department of Emergency Medical Services, Faculty of Health Sciences, Al Qunfudah, Umm Al-Qura University, Makkah 21912, Saudi Arabia;
| | - Masood A. Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia;
| |
Collapse
|
29
|
Rashidi H, Khan I, Dang L, Albahra S, Ratan U, Chadderwala N, To W, Srinivas P, Wajda J, Tran N. Prediction of tuberculosis using an automated machine learning platform for models trained on synthetic data. J Pathol Inform 2022; 13:10. [PMID: 35136677 PMCID: PMC8794034 DOI: 10.4103/jpi.jpi_75_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/18/2021] [Accepted: 11/30/2021] [Indexed: 11/15/2022] Open
Abstract
High-quality medical data is critical to the development and implementation of machine learning (ML) algorithms in healthcare; however, security, and privacy concerns continue to limit access. We sought to determine the utility of “synthetic data” in training ML algorithms for the detection of tuberculosis (TB) from inflammatory biomarker profiles. A retrospective dataset (A) comprised of 278 patients was used to generate synthetic datasets (B, C, and D) for training models prior to secondary validation on a generalization dataset. ML models trained and validated on the Dataset A (real) demonstrated an accuracy of 90%, a sensitivity of 89% (95% CI, 83–94%), and a specificity of 100% (95% CI, 81–100%). Models trained using the optimal synthetic dataset B showed an accuracy of 91%, a sensitivity of 93% (95% CI, 87–96%), and a specificity of 77% (95% CI, 50–93%). Synthetic datasets C and D displayed diminished performance measures (respective accuracies of 71% and 54%). This pilot study highlights the promise of synthetic data as an expedited means for ML algorithm development.
Collapse
|
30
|
Muslu O, Hoyt CT, Lacerda M, Hofmann-Apitius M, Frohlich H. GuiltyTargets: Prioritization of Novel Therapeutic Targets With Network Representation Learning. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:491-500. [PMID: 32750869 DOI: 10.1109/tcbb.2020.3003830] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The majority of clinical trials fail due to low efficacy of investigated drugs, often resulting from a poor choice of target protein. Existing computational approaches aim to support target selection either via genetic evidence or by putting potential targets into the context of a disease specific network reconstruction. The purpose of this work was to investigate whether network representation learning techniques could be used to allow for a machine learning based prioritization of putative targets. We propose a novel target prioritization approach, GuiltyTargets, which relies on attributed network representation learning of a genome-wide protein-protein interaction network annotated with disease-specific differential gene expression and uses positive-unlabeled (PU) machine learning for candidate ranking. We evaluated our approach on 12 datasets from six diseases of different type (cancer, metabolic, neurodegenerative) within a 10 times repeated 5-fold stratified cross-validation and achieved AUROC values between 0.92 - 0.97, significantly outperforming previous approaches that relied on manually engineered topological features. Moreover, we showed that GuiltyTargets allows for target repositioning across related disease areas. An application of GuiltyTargets to Alzheimer's disease resulted in a number of highly ranked candidates that are currently discussed as targets in the literature. Interestingly, one (COMT) is also the target of an approved drug (Tolcapone) for Parkinson's disease, highlighting the potential for target repositioning with our method. The GuiltyTargets Python package is available on PyPI and all code used for analysis can be found under the MIT License at https://github.com/GuiltyTargets. Attributed network representation learning techniques provide an interesting approach to effectively leverage the existing knowledge about the molecular mechanisms in different diseases. In this work, the combination with positive-unlabeled learning for target prioritization demonstrated a clear superiority compared to classical feature engineering approaches. Our work highlights the potential of attributed network representation learning for target prioritization. Given the overarching relevance of networks in computational biology we believe that attributed network representation learning techniques could have a broader impact in the future.
Collapse
|
31
|
Anand S, Iyyappan OR, Manoharan S, Anand D, Jose MA, Shanker RR. Text Mining Protocol to Retrieve Significant Drug-Gene Interactions from PubMed Abstracts. Methods Mol Biol 2022; 2496:17-39. [PMID: 35713857 DOI: 10.1007/978-1-0716-2305-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Genes and proteins form the basis of all cellular processes and ensure a smooth functioning of the human system. The diseases caused in humans can be either genetic in nature or may be caused due to external factors. Genetic diseases are mainly the result of any anomaly in gene/protein structure or function. This disruption interferes with the normal expression of cellular components. Against external factors, even though the immunogenicity of every individual protects them to a certain extent from infections, they are still susceptible to other disease-causing agents. Understanding the biological pathway/entities that could be targeted by specific drugs is an essential component of drug discovery. The traditional drug target discovery process is time-consuming and practically not feasible. A computational approach could provide speed and efficiency to the method. With the presence of vast biomedical literature, text mining also seems to be an obvious choice which could efficiently aid with other computational methods in identifying drug-gene targets. These could aid in initial stages of reviewing the disease components or can even aid parallel in extracting drug-disease-gene/protein relationships from literature. The present chapter aims at finding drug-gene interactions and how the information could be explored for drug interaction.
Collapse
Affiliation(s)
- Sadhanha Anand
- Department of Biomedical Engineering, PSG College of Technology, Coimbatore, Tamilnadu, India
| | - Oviya Ramalakshmi Iyyappan
- Department of Sciences, Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Chennai, Tamilnadu, India
| | - Sharanya Manoharan
- Department of Bioinformatics, Stella Maris College (Autonomous), Chennai, Tamilnadu, India
| | - Dheepa Anand
- Department of Pharmacology, Cheran College of Pharmacy, Coimbatore, Tamilnadu, India
| | | | - Raja Ravi Shanker
- International Business Unit, Alembic Pharmaceuticals Limited, Vadodara, Gujarat, India.
| |
Collapse
|
32
|
Manoharan S, Iyyappan OR. A Hybrid Protocol for Finding Novel Gene Targets for Various Diseases Using Microarray Expression Data Analysis and Text Mining. Methods Mol Biol 2022; 2496:41-70. [PMID: 35713858 DOI: 10.1007/978-1-0716-2305-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The advancement in technology for various scientific experiments and the amount of raw data produced from that is enormous, thus giving rise to various subsets of biologists working with genome, proteome, transcriptome, expression, pathway, and so on. This has led to exponential growth in scientific literature which is becoming beyond the means of manual curation and annotation for extracting information of importance. Microarray data are expression data, analysis of which results in a set of up/downregulated lists of genes that are functionally annotated to ascertain the biological meaning of genes. These genes are represented as vocabularies and/or Gene Ontology terms when associated with pathway enrichment analysis need relational and conceptual understanding to a disease. The chapter deals with a hybrid approach we designed for identifying novel drug-disease targets. Microarray data for muscular dystrophy is explored here as an example and text mining approaches are utilized with an aim to identify promisingly novel drug targets. Our main objective is to give a basic overview from a biologist's perspective for whom text mining approaches of data mining and information retrieval is fairly a new concept. The chapter aims to bridge the gap between biologist and computational text miners and bring about unison for a more informative research in a fast and time efficient manner.
Collapse
Affiliation(s)
- Sharanya Manoharan
- Department of Bioinformatics, Stella Maris College (Autonomous), Chennai, Tamilnadu, India.
| | - Oviya Ramalakshmi Iyyappan
- Department of Sciences, Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Chennai, Tamilnadu, India
| |
Collapse
|
33
|
Selvaraj C, Chandra I, Singh SK. Artificial intelligence and machine learning approaches for drug design: challenges and opportunities for the pharmaceutical industries. Mol Divers 2021; 26:1893-1913. [PMID: 34686947 PMCID: PMC8536481 DOI: 10.1007/s11030-021-10326-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 09/24/2021] [Indexed: 12/27/2022]
Abstract
The global spread of COVID-19 has raised the importance of pharmaceutical drug development as intractable and hot research. Developing new drug molecules to overcome any disease is a costly and lengthy process, but the process continues uninterrupted. The critical point to consider the drug design is to use the available data resources and to find new and novel leads. Once the drug target is identified, several interdisciplinary areas work together with artificial intelligence (AI) and machine learning (ML) methods to get enriched drugs. These AI and ML methods are applied in every step of the computer-aided drug design, and integrating these AI and ML methods results in a high success rate of hit compounds. In addition, this AI and ML integration with high-dimension data and its powerful capacity have taken a step forward. Clinical trials output prediction through the AI/ML integrated models could further decrease the clinical trials cost by also improving the success rate. Through this review, we discuss the backend of AI and ML methods in supporting the computer-aided drug design, along with its challenge and opportunity for the pharmaceutical industry. From the available information or data, the AI and ML based prediction for the high throughput virtual screening. After this integration of AI and ML, the success rate of hit identification has gained a momentum with huge success by providing novel drugs.
Collapse
Affiliation(s)
- Chandrabose Selvaraj
- CADD and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Science Block, Karaikudi, Tamil Nadu, 630004, India.
| | - Ishwar Chandra
- CADD and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Science Block, Karaikudi, Tamil Nadu, 630004, India
| | - Sanjeev Kumar Singh
- CADD and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Science Block, Karaikudi, Tamil Nadu, 630004, India.
| |
Collapse
|
34
|
Mohanty E, Mohanty A. Role of artificial intelligence in peptide vaccine design against RNA viruses. INFORMATICS IN MEDICINE UNLOCKED 2021; 26:100768. [PMID: 34722851 PMCID: PMC8536498 DOI: 10.1016/j.imu.2021.100768] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/16/2021] [Accepted: 10/16/2021] [Indexed: 01/18/2023] Open
Abstract
RNA viruses have high rate of replication and mutation that help them adapt and change according to their environmental conditions. Many viral mutants are the cause of various severe and lethal diseases. Vaccines, on the other hand have the capacity to protect us from infectious diseases by eliciting antibody or cell-mediated immune responses that are pathogen-specific. While there are a few reviews pertaining to the use of artificial intelligence (AI) for SARS-COV-2 vaccine development, none focus on peptide vaccination for RNA viruses and the important role played by AI in it. Peptide vaccine which is slowly coming to be recognized as a safe and effective vaccination strategy has the capacity to overcome the mutant escape problem which is also being currently faced by SARS-COV-2 vaccines in circulation.Here we review the present scenario of peptide vaccines which are developed using mathematical and computational statistics methods to prevent the spread of disease caused by RNA viruses. We also focus on the importance and current stage of AI and mathematical evolutionary modeling using machine learning tools in the establishment of these new peptide vaccines for the control of viral disease.
Collapse
Affiliation(s)
- Eileena Mohanty
- Trident School of Biotech Sciences, Trident Academy of Creative Technology (TACT), Bhubaneswar, Odisha, 751024, India
| | - Anima Mohanty
- School of Biotechnology (KSBT), KIIT University-2, Bhubaneswar, 751024, India
| |
Collapse
|
35
|
Brown SDM. Advances in mouse genetics for the study of human disease. Hum Mol Genet 2021; 30:R274-R284. [PMID: 34089057 PMCID: PMC8490014 DOI: 10.1093/hmg/ddab153] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 01/11/2023] Open
Abstract
The mouse is the pre-eminent model organism for studies of mammalian gene function and has provided an extraordinarily rich range of insights into basic genetic mechanisms and biological systems. Over several decades, the characterization of mouse mutants has illuminated the relationship between gene and phenotype, providing transformational insights into the genetic bases of disease. However, if we are to deliver the promise of genomic and precision medicine, we must develop a comprehensive catalogue of mammalian gene function that uncovers the dark genome and elucidates pleiotropy. Advances in large-scale mouse mutagenesis programmes allied to high-throughput mouse phenomics are now addressing this challenge and systematically revealing novel gene function and multi-morbidities. Alongside the development of these pan-genomic mutational resources, mouse genetics is employing a range of diversity resources to delineate gene-gene and gene-environment interactions and to explore genetic context. Critically, mouse genetics is a powerful tool for assessing the functional impact of human genetic variation and determining the causal relationship between variant and disease. Together these approaches provide unique opportunities to dissect in vivo mechanisms and systems to understand pathophysiology and disease. Moreover, the provision and utility of mouse models of disease has flourished and engages cumulatively at numerous points across the translational spectrum from basic mechanistic studies to pre-clinical studies, target discovery and therapeutic development.
Collapse
|
36
|
Abstract
This review provides the feasible literature on drug discovery through ML tools and techniques that are enforced in every phase of drug development to accelerate the research process and deduce the risk and expenditure in clinical trials. Machine learning techniques improve the decision-making in pharmaceutical data across various applications like QSAR analysis, hit discoveries, de novo drug architectures to retrieve accurate outcomes. Target validation, prognostic biomarkers, digital pathology are considered under problem statements in this review. ML challenges must be applicable for the main cause of inadequacy in interpretability outcomes that may restrict the applications in drug discovery. In clinical trials, absolute and methodological data must be generated to tackle many puzzles in validating ML techniques, improving decision-making, promoting awareness in ML approaches, and deducing risk failures in drug discovery.
Collapse
Affiliation(s)
- Suresh Dara
- Department of Computer Science and Engineering, B V Raju Institute of Technology, Narsapur, Medak, 502313 Telangana India
| | - Swetha Dhamercherla
- Department of Computer Science and Engineering, B V Raju Institute of Technology, Narsapur, Medak, 502313 Telangana India
| | - Surender Singh Jadav
- Centre for Molecular Cancer Research (CMCR) and Vishnu Institute of Pharmaceutical Education and Research (VIPER), Narsapur, Medak, 502313 Telangana India
| | - CH Madhu Babu
- Department of Computer Science and Engineering, B V Raju Institute of Technology, Narsapur, Medak, 502313 Telangana India
| | - Mohamed Jawed Ahsan
- Department of Pharmaceutical Chemistry, Maharishi Arvind College of Pharmacy, Jaipur, 302023 Rajasthan India
| |
Collapse
|
37
|
Serrano Nájera G, Narganes Carlón D, Crowther DJ. TrendyGenes, a computational pipeline for the detection of literature trends in academia and drug discovery. Sci Rep 2021; 11:15747. [PMID: 34344904 PMCID: PMC8333311 DOI: 10.1038/s41598-021-94897-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023] Open
Abstract
Target identification and prioritisation are prominent first steps in modern drug discovery. Traditionally, individual scientists have used their expertise to manually interpret scientific literature and prioritise opportunities. However, increasing publication rates and the wider routine coverage of human genes by omic-scale research make it difficult to maintain meaningful overviews from which to identify promising new trends. Here we propose an automated yet flexible pipeline that identifies trends in the scientific corpus which align with the specific interests of a researcher and facilitate an initial prioritisation of opportunities. Using a procedure based on co-citation networks and machine learning, genes and diseases are first parsed from PubMed articles using a novel named entity recognition system together with publication date and supporting information. Then recurrent neural networks are trained to predict the publication dynamics of all human genes. For a user-defined therapeutic focus, genes generating more publications or citations are identified as high-interest targets. We also used topic detection routines to help understand why a gene is trendy and implement a system to propose the most prominent review articles for a potential target. This TrendyGenes pipeline detects emerging targets and pathways and provides a new way to explore the literature for individual researchers, pharmaceutical companies and funding agencies.
Collapse
Affiliation(s)
- Guillermo Serrano Nájera
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - David Narganes Carlón
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
- Division of Population Health and Genomics, Ninewells Hospital, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
- Exscientia Ltd, Dundee One, River Court, 5 West Victoria Dock Road, Dundee, DD1 3JT, UK
| | - Daniel J Crowther
- Exscientia Ltd, Dundee One, River Court, 5 West Victoria Dock Road, Dundee, DD1 3JT, UK.
| |
Collapse
|
38
|
Hartl D, de Luca V, Kostikova A, Laramie J, Kennedy S, Ferrero E, Siegel R, Fink M, Ahmed S, Millholland J, Schuhmacher A, Hinder M, Piali L, Roth A. Translational precision medicine: an industry perspective. J Transl Med 2021; 19:245. [PMID: 34090480 PMCID: PMC8179706 DOI: 10.1186/s12967-021-02910-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/25/2021] [Indexed: 02/08/2023] Open
Abstract
In the era of precision medicine, digital technologies and artificial intelligence, drug discovery and development face unprecedented opportunities for product and business model innovation, fundamentally changing the traditional approach of how drugs are discovered, developed and marketed. Critical to this transformation is the adoption of new technologies in the drug development process, catalyzing the transition from serendipity-driven to data-driven medicine. This paradigm shift comes with a need for both translation and precision, leading to a modern Translational Precision Medicine approach to drug discovery and development. Key components of Translational Precision Medicine are multi-omics profiling, digital biomarkers, model-based data integration, artificial intelligence, biomarker-guided trial designs and patient-centric companion diagnostics. In this review, we summarize and critically discuss the potential and challenges of Translational Precision Medicine from a cross-industry perspective.
Collapse
Affiliation(s)
- Dominik Hartl
- Novartis Institutes for BioMedical Research, Basel, Switzerland.
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany.
| | - Valeria de Luca
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Anna Kostikova
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Jason Laramie
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Scott Kennedy
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Enrico Ferrero
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Richard Siegel
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Martin Fink
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | | | - Markus Hinder
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Luca Piali
- Roche Innovation Center Basel, Basel, Switzerland
| | - Adrian Roth
- Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
39
|
Vatansever S, Schlessinger A, Wacker D, Kaniskan HÜ, Jin J, Zhou M, Zhang B. Artificial intelligence and machine learning-aided drug discovery in central nervous system diseases: State-of-the-arts and future directions. Med Res Rev 2021; 41:1427-1473. [PMID: 33295676 PMCID: PMC8043990 DOI: 10.1002/med.21764] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/30/2020] [Accepted: 11/20/2020] [Indexed: 01/11/2023]
Abstract
Neurological disorders significantly outnumber diseases in other therapeutic areas. However, developing drugs for central nervous system (CNS) disorders remains the most challenging area in drug discovery, accompanied with the long timelines and high attrition rates. With the rapid growth of biomedical data enabled by advanced experimental technologies, artificial intelligence (AI) and machine learning (ML) have emerged as an indispensable tool to draw meaningful insights and improve decision making in drug discovery. Thanks to the advancements in AI and ML algorithms, now the AI/ML-driven solutions have an unprecedented potential to accelerate the process of CNS drug discovery with better success rate. In this review, we comprehensively summarize AI/ML-powered pharmaceutical discovery efforts and their implementations in the CNS area. After introducing the AI/ML models as well as the conceptualization and data preparation, we outline the applications of AI/ML technologies to several key procedures in drug discovery, including target identification, compound screening, hit/lead generation and optimization, drug response and synergy prediction, de novo drug design, and drug repurposing. We review the current state-of-the-art of AI/ML-guided CNS drug discovery, focusing on blood-brain barrier permeability prediction and implementation into therapeutic discovery for neurological diseases. Finally, we discuss the major challenges and limitations of current approaches and possible future directions that may provide resolutions to these difficulties.
Collapse
Affiliation(s)
- Sezen Vatansever
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Avner Schlessinger
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Daniel Wacker
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - H. Ümit Kaniskan
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jian Jin
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ming‐Ming Zhou
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Bin Zhang
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
40
|
A primer on applying AI synergistically with domain expertise to oncology. Biochim Biophys Acta Rev Cancer 2021; 1876:188548. [PMID: 33901609 DOI: 10.1016/j.bbcan.2021.188548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The concurrent growth of large-scale oncology data alongside the computational methods with which to analyze and model it has created a promising environment for revolutionizing cancer diagnosis, treatment, prevention, and drug discovery. Computational methods applied to large datasets have accelerated the drug discovery process by reducing bottlenecks and widening the search space beyond what is experimentally tractable. As the research community gains understanding of the myriad genetic underpinnings of cancer via sequencing, imaging, screens, and more that are ingested, transformed, and modeled by top open-source machine learning and artificial intelligence tools readily available, the next big drug candidate might seem merely an "Enter" key away. Of course, the reality is more convoluted, but still promising. SCOPE OF REVIEW We present methods to approach the process of building an AI model, with strong emphasis on the aspects of model development we believe to be crucial to success but that are not commonly discussed: diligence in posing questions, identifying suitable datasets and curating them, and collaborating closely with biology and oncology experts while designing and evaluating the model. Digital pathology, Electronic Health Records, and other data types outside of high-throughput molecular data are reviewed well by others and outside of the scope of this review. This review emphasizes the importance of considering the limitations of the datasets, computational methods, and our minds when designing AI models. For example, datasets can be biased towards areas of research interest, funding, and particular patient populations. Neural networks may learn representations and correlations within the data that are grounded not in biological phenomena, but statistical anomalies erroneously extracted from the training data. Researchers may mis-interpret or over-interpret the output, or design and evaluate the training process such that the resultant model generalizes poorly. Fortunately, awareness of the strengths and limitations of applying data analytics and AI to drug discovery enables us to leverage them carefully and insightfully while maximizing their utility. These applications when performed in close collaboration with domain experts, together with continuous critical evaluation, generation of new data to minimize known blind spots as they are found, and rigorous experimental validation, increases the success rate of the study. We will discuss applications including AI-assisted target identification, drug repurposing, patient stratification, and gene prioritization. MAJOR CONCLUSIONS Data analytics and AI have demonstrated capabilities to revolutionize cancer research, prevention, and treatment by maximizing our understanding and use of the expanding panoply of experimental data. However, to separate promise from true utility, computational tools must be carefully designed, critically evaluated, and constantly improved. Once that is achieved, a human-computer hybrid discovery process will outperform one driven by each alone. GENERAL SIGNIFICANCE This review highlights the challenges and promise of synergizing predictive AI models with human expertise towards greater understanding of cancer.
Collapse
|
41
|
Kim H, Kim E, Lee I, Bae B, Park M, Nam H. Artificial Intelligence in Drug Discovery: A Comprehensive Review of Data-driven and Machine Learning Approaches. BIOTECHNOL BIOPROC E 2021; 25:895-930. [PMID: 33437151 PMCID: PMC7790479 DOI: 10.1007/s12257-020-0049-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/27/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023]
Abstract
As expenditure on drug development increases exponentially, the overall drug discovery process requires a sustainable revolution. Since artificial intelligence (AI) is leading the fourth industrial revolution, AI can be considered as a viable solution for unstable drug research and development. Generally, AI is applied to fields with sufficient data such as computer vision and natural language processing, but there are many efforts to revolutionize the existing drug discovery process by applying AI. This review provides a comprehensive, organized summary of the recent research trends in AI-guided drug discovery process including target identification, hit identification, ADMET prediction, lead optimization, and drug repositioning. The main data sources in each field are also summarized in this review. In addition, an in-depth analysis of the remaining challenges and limitations will be provided, and proposals for promising future directions in each of the aforementioned areas.
Collapse
Affiliation(s)
- Hyunho Kim
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Korea
| | - Eunyoung Kim
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Korea
| | - Ingoo Lee
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Korea
| | - Bongsung Bae
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Korea
| | - Minsu Park
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Korea
| | - Hojung Nam
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Korea
| |
Collapse
|
42
|
Yu K, Visweswaran S, Batmanghelich K. Semi-supervised Hierarchical Drug Embedding in Hyperbolic Space. J Chem Inf Model 2020; 60:5647-5657. [PMID: 33140969 PMCID: PMC7943198 DOI: 10.1021/acs.jcim.0c00681] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Learning accurate drug representations is essential for tasks such as computational drug repositioning and prediction of drug side effects. A drug hierarchy is a valuable source that encodes knowledge of relations among drugs in a tree-like structure where drugs that act on the same organs, treat the same disease, or bind to the same biological target are grouped together. However, its utility in learning drug representations has not yet been explored, and currently described drug representations cannot place novel molecules in a drug hierarchy. Here, we develop a semi-supervised drug embedding that incorporates two sources of information: (1) underlying chemical grammar that is inferred from chemical structures of drugs and drug-like molecules (unsupervised) and (2) hierarchical relations that are encoded in an expert-crafted hierarchy of approved drugs (supervised). We use the Variational Auto-Encoder (VAE) framework to encode the chemical structures of molecules and use the drug-drug similarity information obtained from the hierarchy to induce the clustering of drugs in hyperbolic space. The hyperbolic space is amenable for encoding hierarchical relations. Both quantitative and qualitative results support that the learned drug embedding can accurately reproduce the chemical structure and recapitulate the hierarchical relations among drugs. Furthermore, our approach can infer the pharmacological properties of novel molecules by retrieving similar drugs from the embedding space. We demonstrate that our drug embedding can predict new uses and discover new side effects of existing drugs. We show that it significantly outperforms comparison methods in both tasks.
Collapse
Affiliation(s)
- Ke Yu
- Intelligent Systems Program, School of Computing and Information, University of Pittsburgh, Pittsburgh, Pennsylvania 15206, United States
| | - Shyam Visweswaran
- Intelligent Systems Program, School of Computing and Information, University of Pittsburgh, Pittsburgh, Pennsylvania 15206, United States
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania 15206, United States
| | - Kayhan Batmanghelich
- Intelligent Systems Program, School of Computing and Information, University of Pittsburgh, Pittsburgh, Pennsylvania 15206, United States
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania 15206, United States
| |
Collapse
|
43
|
Chen Z, Liu X, Hogan W, Shenkman E, Bian J. Applications of artificial intelligence in drug development using real-world data. Drug Discov Today 2020; 26:1256-1264. [PMID: 33358699 DOI: 10.1016/j.drudis.2020.12.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/21/2020] [Accepted: 12/16/2020] [Indexed: 01/12/2023]
Abstract
The US Food and Drug Administration (FDA) has been actively promoting the use of real-world data (RWD) in drug development. RWD can generate important real-world evidence reflecting the real-world clinical environment where the treatments are used. Meanwhile, artificial intelligence (AI), especially machine- and deep-learning (ML/DL) methods, have been increasingly used across many stages of the drug development process. Advancements in AI have also provided new strategies to analyze large, multidimensional RWD. Thus, we conducted a rapid review of articles from the past 20 years, to provide an overview of the drug development studies that use both AI and RWD. We found that the most popular applications were adverse event detection, trial recruitment, and drug repurposing. Here, we also discuss current research gaps and future opportunities.
Collapse
Affiliation(s)
- Zhaoyi Chen
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32610-0177, USA
| | - Xiong Liu
- AI Innovation Center, Novartis, Cambridge, MA 02142, USA
| | - William Hogan
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32610-0177, USA
| | - Elizabeth Shenkman
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32610-0177, USA
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32610-0177, USA.
| |
Collapse
|
44
|
Artificial intelligence in the early stages of drug discovery. Arch Biochem Biophys 2020; 698:108730. [PMID: 33347838 DOI: 10.1016/j.abb.2020.108730] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
Although the use of computational methods within the pharmaceutical industry is well established, there is an urgent need for new approaches that can improve and optimize the pipeline of drug discovery and development. In spite of the fact that there is no unique solution for this need for innovation, there has recently been a strong interest in the use of Artificial Intelligence for this purpose. As a matter of fact, not only there have been major contributions from the scientific community in this respect, but there has also been a growing partnership between the pharmaceutical industry and Artificial Intelligence companies. Beyond these contributions and efforts there is an underlying question, which we intend to discuss in this review: can the intrinsic difficulties within the drug discovery process be overcome with the implementation of Artificial Intelligence? While this is an open question, in this work we will focus on the advantages that these algorithms provide over the traditional methods in the context of early drug discovery.
Collapse
|
45
|
Fahimian G, Zahiri J, Arab SS, Sajedi RH. RepCOOL: computational drug repositioning via integrating heterogeneous biological networks. J Transl Med 2020; 18:375. [PMID: 33008415 PMCID: PMC7532104 DOI: 10.1186/s12967-020-02541-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 09/21/2020] [Indexed: 01/07/2023] Open
Abstract
Background It often takes more than 10 years and costs more than 1 billion dollars to develop a new drug for a particular disease and bring it to the market. Drug repositioning can significantly reduce costs and time in drug development. Recently, computational drug repositioning attracted a considerable amount of attention among researchers, and a plethora of computational drug repositioning methods have been proposed. This methodology has widely been used in order to address various medical challenges, including cancer treatment. The most common cancers are lung and breast cancers. Thus, suggesting FDA-approved drugs via drug repositioning for breast cancer would help us to circumvent the approval process and subsequently save money as well as time. Methods In this study, we propose a novel network-based method, named RepCOOL, for drug repositioning. RepCOOL integrates various heterogeneous biological networks to suggest new drug candidates for a given disease. Results The proposed method showed a promising performance on benchmark datasets via rigorous cross-validation. The final drug repositioning model has been built based on a random forest classifier after examining various machine learning algorithms. Finally, in a case study, four FDA approved drugs were suggested for breast cancer stage II. Conclusion Results show the potency of the proposed method in detecting true drug-disease relationships. RepCOOL suggested four new drugs for breast cancer stage II namely Doxorubicin, Paclitaxel, Trastuzumab, and Tamoxifen.
Collapse
Affiliation(s)
- Ghazale Fahimian
- Bioinformatics and Computational Omics Lab (BioCOOL), Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javad Zahiri
- Bioinformatics and Computational Omics Lab (BioCOOL), Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Seyed Shahriar Arab
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza H Sajedi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
46
|
Raschka S, Kaufman B. Machine learning and AI-based approaches for bioactive ligand discovery and GPCR-ligand recognition. Methods 2020; 180:89-110. [PMID: 32645448 PMCID: PMC8457393 DOI: 10.1016/j.ymeth.2020.06.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/23/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
In the last decade, machine learning and artificial intelligence applications have received a significant boost in performance and attention in both academic research and industry. The success behind most of the recent state-of-the-art methods can be attributed to the latest developments in deep learning. When applied to various scientific domains that are concerned with the processing of non-tabular data, for example, image or text, deep learning has been shown to outperform not only conventional machine learning but also highly specialized tools developed by domain experts. This review aims to summarize AI-based research for GPCR bioactive ligand discovery with a particular focus on the most recent achievements and research trends. To make this article accessible to a broad audience of computational scientists, we provide instructive explanations of the underlying methodology, including overviews of the most commonly used deep learning architectures and feature representations of molecular data. We highlight the latest AI-based research that has led to the successful discovery of GPCR bioactive ligands. However, an equal focus of this review is on the discussion of machine learning-based technology that has been applied to ligand discovery in general and has the potential to pave the way for successful GPCR bioactive ligand discovery in the future. This review concludes with a brief outlook highlighting the recent research trends in deep learning, such as active learning and semi-supervised learning, which have great potential for advancing bioactive ligand discovery.
Collapse
Affiliation(s)
- Sebastian Raschka
- University of Wisconsin-Madison, Department of Statistics, United States.
| | - Benjamin Kaufman
- University of Wisconsin-Madison, Department of Biostatistics and Medical Informatics, United States
| |
Collapse
|
47
|
Bazaga A, Leggate D, Weisser H. Genome-wide investigation of gene-cancer associations for the prediction of novel therapeutic targets in oncology. Sci Rep 2020; 10:10787. [PMID: 32612205 PMCID: PMC7330039 DOI: 10.1038/s41598-020-67846-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/08/2020] [Indexed: 02/03/2023] Open
Abstract
A major cause of failed drug discovery programs is suboptimal target selection, resulting in the development of drug candidates that are potent inhibitors, but ineffective at treating the disease. In the genomics era, the availability of large biomedical datasets with genome-wide readouts has the potential to transform target selection and validation. In this study we investigate how computational intelligence methods can be applied to predict novel therapeutic targets in oncology.
We compared different machine learning classifiers applied to the task of drug target classification for nine different human cancer types. For each cancer type, a set of “known” target genes was obtained and equally-sized sets of “non-targets” were sampled multiple times from the human protein-coding genes. Models were trained on mutation, gene expression (TCGA), and gene essentiality (DepMap) data. In addition, we generated a numerical embedding of the interaction network of protein-coding genes using deep network representation learning and included the results in the modeling. We assessed feature importance using a random forests classifier and performed feature selection based on measuring permutation importance against a null distribution. Our best models achieved good generalization performance based on the AUROC metric. With the best model for each cancer type, we ran predictions on more than 15,000 protein-coding genes to identify potential novel targets. Our results indicate that this approach may be useful to inform early stages of the drug discovery pipeline.
Collapse
Affiliation(s)
- Adrián Bazaga
- Department of Genetics, University of Cambridge, Cambridge, UK. .,STORM Therapeutics Ltd, Cambridge, UK.
| | | | | |
Collapse
|
48
|
Wang Y, Chen Z, Bian F, Shang L, Zhu K, Zhao Y. Advances of droplet-based microfluidics in drug discovery. Expert Opin Drug Discov 2020; 15:969-979. [DOI: 10.1080/17460441.2020.1758663] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Yuetong Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zhuoyue Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Feika Bian
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Luoran Shang
- Zhongshan-Xuhui Hospital, Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Kaixuan Zhu
- School of Electrical and Information Engineering, Suzhou Institute of Technology, Jiangsu University of Science and Technology, Zhangjiagang, China
| | - Yuanjin Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| |
Collapse
|
49
|
Liu E, Zhang ZZ, Cheng X, Liu X, Cheng L. SCNrank: spectral clustering for network-based ranking to reveal potential drug targets and its application in pancreatic ductal adenocarcinoma. BMC Med Genomics 2020; 13:50. [PMID: 32241274 PMCID: PMC7119297 DOI: 10.1186/s12920-020-0681-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is the most common pancreatic malignancy. Due to its wide heterogeneity, PDAC acts aggressively and responds poorly to most chemotherapies, causing an urgent need for the development of new therapeutic strategies. Cell lines have been used as the foundation for drug development and disease modeling. CRISPR-Cas9 plays a key role in every step-in drug discovery: from target identification and validation to preclinical cancer cell testing. Using cell-line models and CRISPR-Cas9 technology together make drug target prediction feasible. However, there is still a large gap between predicted results and actionable targets in real tumors. Biological network models provide great modus to mimic genetic interactions in real biological systems, which can benefit gene perturbation studies and potential target identification for treating PDAC. Nevertheless, building a network model that takes cell-line data and CRISPR-Cas9 data as input to accurately predict potential targets that will respond well on real tissue remains unsolved. Methods We developed a novel algorithm ‘Spectral Clustering for Network-based target Ranking’ (SCNrank) that systematically integrates three types of data: expression profiles from tumor tissue, normal tissue and cell-line PDAC; protein-protein interaction network (PPI); and CRISPR-Cas9 data to prioritize potential drug targets for PDAC. The whole algorithm can be classified into three steps: 1. using STRING PPI network skeleton, SCNrank constructs tissue-specific networks with PDAC tumor and normal pancreas tissues from expression profiles; 2. With the same network skeleton, SCNrank constructs cell-line-specific networks using the cell-line PDAC expression profiles and CRISPR-Cas 9 data from pancreatic cancer cell-lines; 3. SCNrank applies a novel spectral clustering approach to reduce data dimension and generate gene clusters that carry common features from both networks. Finally, SCNrank applies a scoring scheme called ‘Target Influence score’ (TI), which estimates a given target’s influence towards the cluster it belongs to, for scoring and ranking each drug target. Results We applied SCNrank to analyze 263 expression profiles, CRPSPR-Cas9 data from 22 different pancreatic cancer cell-lines and the STRING protein-protein interaction (PPI) network. With SCNrank, we successfully constructed an integrated tissue PDAC network and an integrated cell-line PDAC network, both of which contain 4414 selected genes that are overexpressed in tumor tissue samples. After clustering, 4414 genes are distributed into 198 clusters, which include 367 targets of FDA approved drugs. These drug targets are all scored and ranked by their TI scores, which we defined to measure their influence towards the network. We validated top-ranked targets in three aspects: Firstly, mapping them onto the existing clinical drug targets of PDAC to measure the concordance. Secondly, we performed enrichment analysis to these drug targets and the clusters there are within, to reveal functional associations between clusters and PDAC; Thirdly, we performed survival analysis for the top-ranked targets to connect targets with clinical outcomes. Survival analysis reveals that overexpression of three top-ranked genes, PGK1, HMMR and POLE2, significantly increases the risk of death in PDAC patients. Conclusion SCNrank is an unbiased algorithm that systematically integrates multiple types of omics data to do potential drug target selection and ranking. SCNrank shows great capability in predicting drug targets for PDAC. Pancreatic cancer-associated gene candidates predicted by our SCNrank approach have the potential to guide genetics-based anti-pancreatic drug discovery.
Collapse
Affiliation(s)
- Enze Liu
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University-Purdue University, Indianapolis, IN, 46202, USA
| | - Zhuang Zhuang Zhang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Xiaolin Cheng
- College of Pharmacy, Division of Medicinal Chemistry and Pharmacognosy, the Ohio State University, Columbus, OH, 43210, USA
| | - Xiaoqi Liu
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA.
| | - Lijun Cheng
- Department of Biomedical informatics, College of medicine, the Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
50
|
Réda C, Kaufmann E, Delahaye-Duriez A. Machine learning applications in drug development. Comput Struct Biotechnol J 2019; 18:241-252. [PMID: 33489002 PMCID: PMC7790737 DOI: 10.1016/j.csbj.2019.12.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023] Open
Abstract
Due to the huge amount of biological and medical data available today, along with well-established machine learning algorithms, the design of largely automated drug development pipelines can now be envisioned. These pipelines may guide, or speed up, drug discovery; provide a better understanding of diseases and associated biological phenomena; help planning preclinical wet-lab experiments, and even future clinical trials. This automation of the drug development process might be key to the current issue of low productivity rate that pharmaceutical companies currently face. In this survey, we will particularly focus on two classes of methods: sequential learning and recommender systems, which are active biomedical fields of research.
Collapse
Affiliation(s)
- Clémence Réda
- NeuroDiderot, UMR 1141, Inserm, Université de Paris, Sorbonne Paris Cité, Hôpital Robert Debré, 48, boulevard Sérurier, Paris 75019, France
- Université Paris Diderot, Université de Paris, Sorbonne Paris Cité, 5, rue Thomas Mann, Paris 75013, France
| | - Emilie Kaufmann
- Univ. Lille, CNRS, Centrale Lille, Inria, UMR 9189 - CRIStAL - Centre de Recherche en Informatique Signal et Automatique de Lille, F-59000 Lille, France
| | - Andrée Delahaye-Duriez
- NeuroDiderot, UMR 1141, Inserm, Université de Paris, Sorbonne Paris Cité, Hôpital Robert Debré, 48, boulevard Sérurier, Paris 75019, France
- Université Paris 13, Sorbonne Paris Cité, UFR de santé, médecine et biologie humaine, Bobigny 93000, France
- Service histologie-embryologie-cytogénétique-biologie de la reproduction-CECOS, Hôpital Jean Verdier, AP-HP, Bondy 93140, France
| |
Collapse
|