1
|
Provasi D, Filizola M. Fine-Tuned Deep Transfer Learning Models for Large Screenings of Safer Drugs Targeting Class A GPCRs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.07.627102. [PMID: 39713468 PMCID: PMC11661127 DOI: 10.1101/2024.12.07.627102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
G protein-coupled receptors (GPCRs) remain a focal point of research due to their critical roles in cell signaling and their prominence as drug targets. However, directly linking drug efficacy to receptor-mediated activation of specific intracellular transducers and the resulting physiological outcomes remains challenging. It is unclear whether the enhanced therapeutic window of certain drugs - defined as the dose range that provides effective therapy with minimal side effects - stems from their low intrinsic efficacy across all signaling pathways or ligand bias, wherein specific transducer subtypes are preferentially activated in a given cellular system compared to a reference ligand. Accurately predicting safer compounds, whether through low intrinsic efficacy or ligand bias, would greatly advance drug development. While AI models hold promise for such predictions, the development of deep learning models capable of reliably forecasting GPCR ligands with defined bioactivities remains challenging, largely due to the limited availability of high-quality data. To address this, we pre-trained a model on receptor sequences and ligand datasets across all class A GPCRs, and then refined it to predict low-efficacy compounds or biased agonists for individual class A GPCRs. This was achieved using transfer learning and a neural network incorporating natural language processing of target sequences and receptor mutation effects on signaling. These two fine-tuned models-one for low-efficacy agonists and one for biased agonists-are available on demand for each class A GPCR and enable virtual screening of large chemical libraries, thereby facilitating the discovery of compounds with potentially improved safety profiles.
Collapse
|
2
|
Dhanabalan AK, Devadasan V, Haribabu J, Krishnasamy G. Machine learning models to identify lead compound and substitution optimization to have derived energetics and conformational stability through docking and MD simulations for sphingosine kinase 1. Mol Divers 2024:10.1007/s11030-024-10997-4. [PMID: 39417979 DOI: 10.1007/s11030-024-10997-4] [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: 08/14/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Sphingosine kinases (SphKs) are a group of important enzymes that circulate at low micromolar concentrations in mammals and have received considerable attention due to the roles they play in a broad array of biological processes including apoptosis, mutagenesis, lymphocyte migration, radio- and chemo-sensitization, and angiogenesis. In the present study, we constructed three classification models by four machine learning (ML) algorithms including naive bayes (NB), support vector machine (SVM), logistic regression, and random forest from 395 compounds. The generated ML models were validated by fivefold cross validation. Five different scaffold hit fragments resulted from SVM model-based virtual screening and docking results indicate that all the five fragments exhibit common hydrogen bond interaction a catalytic residue of SphK1. Further, molecular dynamics (MD) simulations and binding free energy calculation had been carried out with the identified five fragment leads and three cocrystal inhibitors. The best 15 fragments were selected. Molecular dynamics (MD) simulations showed that among these compounds, 7 compounds have favorable binding energy compared with cocrystal inhibitors. Hence, the study showed that the present lead fragments could act as potential inhibitors against therapeutic target of cancers and neurodegenerative disorders.
Collapse
Affiliation(s)
- Anantha Krishnan Dhanabalan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Velmurugan Devadasan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| | - Jebiti Haribabu
- Facultad de Medicina, Universidad de Atacama, Los Carreras 1579, 1532502, Copiapó, Chile
- Chennai Institute of Technology (CIT), Chennai, Tamil Nadu, 600069, India
| | - Gunasekaran Krishnasamy
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai, Tamil Nadu, 600025, India.
| |
Collapse
|
3
|
Wang Z, Huo M, Qiao L, Qiao Y, Zhang Y. SYSTCM: A systemic web platform for objective identification of pharmacological effects based on interplay of "traditional Chinese Medicine-components-targets". Comput Biol Med 2024; 179:108878. [PMID: 39043107 DOI: 10.1016/j.compbiomed.2024.108878] [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: 02/07/2024] [Revised: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
Mechanism analysis is essential for the use and promotion of Traditional Chinese Medicine (TCM). Traditional methods of network analysis relying on expert experience lack an explanatory framework, prompting the application of deep learning and machine learning for objective identification of TCM pharmacological effects. A dataset was used to construct an interacted network graph between 424 molecular descriptors and 465 pharmacological targets to represent the relationship between components and pharmacological effects. Subsequently, the optimal identification model of pharmacological effects (IPE) was established through convolution neural networks of GoogLeNet structure. The AUC values are greater than 0.8, MCC values are greater than 0.7, and ACC values are greater than 0.85 across various test datasets. Subsequently, 18 recognition models of TCM efficacy (RTE) were created using support vector machines (SVM). Integration of pharmacological effects and efficacies led to the development of the systemic web platform for identification of pharmacological effects (SYSTCM). The platform, comprising 70,961 terms, including 636 Traditional Chinese Medicines (TCMs), 8190 components, 40 pharmacological effects, and 18 efficacies. Through the SYSTCM platform, (1) Total 100 components were predicted from TCMs with anti-inflammatory pharmacological effects. (2) The pharmacological effects of complete constituents were predicted from Coptidis Rhizoma (Huang Lian). (3) The principal components, pharmacological effects, and efficacies were elucidated from Salviae Miltiorrhizae radix et rhizome (Dan Shen). SYSTCM addresses subjectivity in pharmacological effect determination, offering a potential avenue for advancing TCM drug development and clinical applications. Access SYSTCM at http://systcm.cn.
Collapse
Affiliation(s)
- Zewen Wang
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Mengqi Huo
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Liansheng Qiao
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yanjiang Qiao
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Yanling Zhang
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
4
|
Pawar SB, Deshmukh NK, Jadhav SB. Hybrid deep learning technique for COX-2 inhibition bioactivity detection against breast cancer disease. Biomed Eng Lett 2024; 14:631-647. [PMID: 39512384 PMCID: PMC11538098 DOI: 10.1007/s13534-024-00355-6] [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: 06/16/2023] [Revised: 01/03/2024] [Accepted: 01/24/2024] [Indexed: 11/15/2024] Open
Abstract
This study addresses detecting COX-2 inhibition in breast cancer, targeting its role in tumor growth. The primary goal is to develop an efficient technique for precise COX-2 inhibition bioactivity detection, with implications for identifying anti-cancer compounds and advancing breast cancer therapies. The proposed methodology uses the UNet architecture for feature extraction, enhancing accuracy. A modified chicken swarm optimization (MCSO) algorithm addresses data dimensionality, optimizing features. An improved Laguerre neural network (ILNN) classifies COX-2 inhibition bioactivity. Validation is performed using the ChEMBL database. The research evaluates the accuracy, precision, recall, F-measure, Matthews' correlation coefficient (MCC), and Dice coefficient of the proposed method. These metrics are compared against those of contemporary methods to assess the efficiency and effectiveness of the developed technique. The study underscores the hybrid deep learning method's significance in accurately detecting COX-2 inhibition bioactivity against breast cancer. Results highlight its potential as a valuable tool in breast cancer drug discovery.
Collapse
Affiliation(s)
- Sahebrao B. Pawar
- School of Computational Sciences, Swami Ramanand Teerth, Marathvada University, Nanded, India
| | - N. K. Deshmukh
- School of Computational Sciences, Swami Ramanand Teerth, Marathvada University, Nanded, India
| | - Sharad B. Jadhav
- School of Computational Sciences, Swami Ramanand Teerth, Marathvada University, Nanded, India
| |
Collapse
|
5
|
Nguyen ATN, Nguyen DTN, Koh HY, Toskov J, MacLean W, Xu A, Zhang D, Webb GI, May LT, Halls ML. The application of artificial intelligence to accelerate G protein-coupled receptor drug discovery. Br J Pharmacol 2024; 181:2371-2384. [PMID: 37161878 DOI: 10.1111/bph.16140] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 04/14/2023] [Accepted: 04/27/2023] [Indexed: 05/11/2023] Open
Abstract
The application of artificial intelligence (AI) approaches to drug discovery for G protein-coupled receptors (GPCRs) is a rapidly expanding area. Artificial intelligence can be used at multiple stages during the drug discovery process, from aiding our understanding of the fundamental actions of GPCRs to the discovery of new ligand-GPCR interactions or the prediction of clinical responses. Here, we provide an overview of the concepts behind artificial intelligence, including the subfields of machine learning and deep learning. We summarise the published applications of artificial intelligence to different stages of the GPCR drug discovery process. Finally, we reflect on the benefits and limitations of artificial intelligence and share our vision for the exciting potential for further development of applications to aid GPCR drug discovery. In addition to making the drug discovery process "faster, smarter and cheaper," we anticipate that the application of artificial intelligence will create exciting new opportunities for GPCR drug discovery. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Anh T N Nguyen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Diep T N Nguyen
- Department of Information Technology, Faculty of Engineering and Technology, Vietnam National University, Cau Giay, Hanoi, Vietnam
| | - Huan Yee Koh
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Monash Data Futures Institute and Department of Data Science and Artificial Intelligence, Monash University, Clayton, Victoria, Australia
| | - Jason Toskov
- Monash DeepNeuron, Monash University, Clayton, Victoria, Australia
| | - William MacLean
- Monash DeepNeuron, Monash University, Clayton, Victoria, Australia
| | - Andrew Xu
- Monash DeepNeuron, Monash University, Clayton, Victoria, Australia
| | - Daokun Zhang
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Monash Data Futures Institute and Department of Data Science and Artificial Intelligence, Monash University, Clayton, Victoria, Australia
| | - Geoffrey I Webb
- Monash Data Futures Institute and Department of Data Science and Artificial Intelligence, Monash University, Clayton, Victoria, Australia
| | - Lauren T May
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
6
|
Ahmad S, Raza K. An extensive review on lung cancer therapeutics using machine learning techniques: state-of-the-art and perspectives. J Drug Target 2024; 32:635-646. [PMID: 38662768 DOI: 10.1080/1061186x.2024.2347358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
There are over 100 types of human cancer, accounting for millions of deaths every year. Lung cancer alone claims over 1.8 million lives per year and is expected to surpass 3.2 million by 2050, which underscores the urgent need for rapid drug development and repurposing initiatives. The application of AI emerges as a pivotal solution to developing anti-cancer therapeutics. This state-of-the-art review aims to explore the various applications of AI in lung cancer therapeutics. Predictive models can analyse large datasets, including clinical data, genetic information, and treatment outcomes, for novel drug design and to generate personalised treatment recommendations, potentially optimising therapeutic strategies, enhancing treatment efficacy, and minimising adverse effects. A thorough literature review study was conducted based on articles indexed in PubMed and Scopus. We compiled the use of various machine learning approaches, including CNN, RNN, GAN, VAEs, and other AI techniques, enhancing efficiency with accuracy exceeding 95%, which is validated through a computer-aided drug design process. AI can revolutionise lung cancer therapeutics, streamlining processes and saving biological scientists' time and effort-however, further research is needed to overcome challenges and fully unlock AI's potential in Lung Cancer Therapeutics.
Collapse
Affiliation(s)
- Shaban Ahmad
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Khalid Raza
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
7
|
Qiu Y, Hou Y, Gohel D, Zhou Y, Xu J, Bykova M, Yang Y, Leverenz JB, Pieper AA, Nussinov R, Caldwell JZK, Brown JM, Cheng F. Systematic characterization of multi-omics landscape between gut microbial metabolites and GPCRome in Alzheimer's disease. Cell Rep 2024; 43:114128. [PMID: 38652661 DOI: 10.1016/j.celrep.2024.114128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/06/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024] Open
Abstract
Shifts in the magnitude and nature of gut microbial metabolites have been implicated in Alzheimer's disease (AD), but the host receptors that sense and respond to these metabolites are largely unknown. Here, we develop a systems biology framework that integrates machine learning and multi-omics to identify molecular relationships of gut microbial metabolites with non-olfactory G-protein-coupled receptors (termed the "GPCRome"). We evaluate 1.09 million metabolite-protein pairs connecting 408 human GPCRs and 335 gut microbial metabolites. Using genetics-derived Mendelian randomization and integrative analyses of human brain transcriptomic and proteomic profiles, we identify orphan GPCRs (i.e., GPR84) as potential drug targets in AD and that triacanthine experimentally activates GPR84. We demonstrate that phenethylamine and agmatine significantly reduce tau hyperphosphorylation (p-tau181 and p-tau205) in AD patient induced pluripotent stem cell-derived neurons. This study demonstrates a systems biology framework to uncover the GPCR targets of human gut microbiota in AD and other complex diseases if broadly applied.
Collapse
Affiliation(s)
- Yunguang Qiu
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yuan Hou
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dhruv Gohel
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yadi Zhou
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jielin Xu
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Marina Bykova
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yuxin Yang
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Andrew A Pieper
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Jessica Z K Caldwell
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA; Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Las Vegas, NV 89106, USA
| | - J Mark Brown
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA; Department of Cancer Biology, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44195, USA; Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA; Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
8
|
Janero DR. Current strategic trends in drug discovery: the present as prologue. Expert Opin Drug Discov 2024; 19:147-159. [PMID: 37936504 DOI: 10.1080/17460441.2023.2275640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023]
Abstract
INTRODUCTION Escalating costs and inherent uncertainties associated with drug discovery invite initiatives to improve its efficiency and de-risk campaigns for inventing better therapeutics. One such initiative involves recognizing and exploiting current approaches in therapeutics invention with molecular mechanisms of action that hold promise for designing and targeting new chemical entities as drugs. AREAS COVERED This perspective considers the current contextual framework around three drug-discovery approaches and evaluates their potential to help identify new targets/modalities in small-molecule molecular pharmacology: diversifying ligand-directed phenotypes for G protein-coupled receptor (GPCR) pharmacotherapeutic signaling; developing therapeutic-protein degraders and stabilizers for proximity-inducing pharmacology; and mining organelle biology for druggable therapeutic targets. EXPERT OPINION The contemporary drug-discovery approaches examined appear generalizable and versatile to have applications in therapeutics invention beyond those case studies discussed herein. Accordingly, they may be considered strategic trends worthy of note in advancing the field toward novel ways of addressing pharmacotherapeutically unmet medical needs.
Collapse
Affiliation(s)
- David R Janero
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, and Health Sciences Entrepreneurs, Northeastern University, Boston, MA, USA
| |
Collapse
|
9
|
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
|
10
|
Provasi D, Filizola M. Enhancing Opioid Bioactivity Predictions through Integration of Ligand-Based and Structure-Based Drug Discovery Strategies with Transfer and Deep Learning Techniques. J Phys Chem B 2023; 127:10691-10699. [PMID: 38084046 PMCID: PMC11252170 DOI: 10.1021/acs.jpcb.3c05306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
The opioid epidemic has cast a shadow over public health, necessitating immediate action to address its devastating consequences. To effectively combat this crisis, it is crucial to discover better opioid drugs with reduced addiction potential. Artificial intelligence-based and other machine learning tools, particularly deep learning models, have garnered significant attention in recent years for their potential to advance drug discovery. However, using these tools poses challenges, especially when training samples are insufficient to achieve adequate prediction performance. In this study, we investigate the effectiveness of transfer learning in building robust deep learning models to enhance ligand bioactivity prediction for each individual opioid receptor (OR) subtype. This is achieved by leveraging knowledge obtained from pretraining a model using supervised learning on a larger data set of bioactivity data combined with ligand-based and structure-based molecular descriptors related to the entire OR subfamily. Our studies hold the potential to advance opioid research by enabling the rapid identification of novel chemical probes with specific bioactivities, which can aid in the study of receptor function and contribute to the future development of improved opioid therapeutics.
Collapse
Affiliation(s)
- Davide Provasi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York 10029, United States
| | - Marta Filizola
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York 10029, United States
| |
Collapse
|
11
|
Murray JD, Lange JJ, Bennett-Lenane H, Holm R, Kuentz M, O'Dwyer PJ, Griffin BT. Advancing algorithmic drug product development: Recommendations for machine learning approaches in drug formulation. Eur J Pharm Sci 2023; 191:106562. [PMID: 37562550 DOI: 10.1016/j.ejps.2023.106562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/09/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
Artificial intelligence is a rapidly expanding area of research, with the disruptive potential to transform traditional approaches in the pharmaceutical industry, from drug discovery and development to clinical practice. Machine learning, a subfield of artificial intelligence, has fundamentally transformed in silico modelling and has the capacity to streamline clinical translation. This paper reviews data-driven modelling methodologies with a focus on drug formulation development. Despite recent advances, there is limited modelling guidance specific to drug product development and a trend towards suboptimal modelling practices, resulting in models that may not give reliable predictions in practice. There is an overwhelming focus on benchtop experimental outcomes obtained for a specific modelling aim, leaving the capabilities of data scraping or the use of combined modelling approaches yet to be fully explored. Moreover, the preference for high accuracy can lead to a reliance on black box methods over interpretable models. This further limits the widespread adoption of machine learning as black boxes yield models that cannot be easily understood for the purposes of enhancing product performance. In this review, recommendations for conducting machine learning research for drug product development to ensure trustworthiness, transparency, and reliability of the models produced are presented. Finally, possible future directions on how research in this area might develop are discussed to aim for models that provide useful and robust guidance to formulators.
Collapse
Affiliation(s)
- Jack D Murray
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Justus J Lange
- School of Pharmacy, University College Cork, Cork, Ireland; Roche Pharmaceutical Research & Early Development, Pre-Clinical CMC, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel, Switzerland
| | | | - René Holm
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| | - Martin Kuentz
- School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz CH 4132, Switzerland
| | | | | |
Collapse
|
12
|
Viswanathan K, Goel M, Laghuvarapu S, Varma G, Priyakumar UD. Streamlining pipeline efficiency: a novel model-agnostic technique for accelerating conditional generative and virtual screening pipelines. Sci Rep 2023; 13:21069. [PMID: 38030689 PMCID: PMC10686981 DOI: 10.1038/s41598-023-42952-y] [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: 03/07/2023] [Accepted: 09/16/2023] [Indexed: 12/01/2023] Open
Abstract
The discovery of potential therapeutic agents for life-threatening diseases has become a significant problem. There is a requirement for fast and accurate methods to identify drug-like molecules that can be used as potential candidates for novel targets. Existing techniques like high-throughput screening and virtual screening are time-consuming and inefficient. Traditional molecule generation pipelines are more efficient than virtual screening but use time-consuming docking software. Such docking functions can be emulated using Machine Learning models with comparable accuracy and faster execution times. However, we find that when pre-trained machine learning models are employed in generative pipelines as oracles, they suffer from model degradation in areas where data is scarce. In this study, we propose an active learning-based model that can be added as a supplement to enhanced molecule generation architectures. The proposed method uses uncertainty sampling on the molecules created by the generator model and dynamically learns as the generator samples molecules from different regions of the chemical space. The proposed framework can generate molecules with high binding affinity with [Formula: see text]a 70% improvement in runtime compared to the baseline model by labeling only [Formula: see text]30% of molecules compared to the baseline oracle.
Collapse
Affiliation(s)
- Karthik Viswanathan
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad, 500032, India
| | - Manan Goel
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad, 500032, India
| | - Siddhartha Laghuvarapu
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad, 500032, India
| | - Girish Varma
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad, 500032, India
| | - U Deva Priyakumar
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad, 500032, India.
| |
Collapse
|
13
|
Liu W, Hopkins AM, Yan P, Du S, Luyt LG, Li Y, Hou J. Can machine learning 'transform' peptides/peptidomimetics into small molecules? A case study with ghrelin receptor ligands. Mol Divers 2023; 27:2239-2255. [PMID: 36331785 DOI: 10.1007/s11030-022-10555-w] [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: 09/06/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
There has been considerable interest in transforming peptides into small molecules as peptide-based molecules often present poorer bioavailability and lower metabolic stability. Our studies looked into building machine learning (ML) models to investigate if ML is able to identify the 'bioactive' features of peptides and use the features to accurately discriminate between binding and non-binding small molecules. The ghrelin receptor (GR), a receptor that is implicated in various diseases, was used as an example to demonstrate whether ML models derived from a peptide library can be used to predict small molecule binders. ML models based on three different algorithms, namely random forest, support vector machine, and extreme gradient boosting, were built based on a carefully curated dataset of peptide/peptidomimetic and small molecule GR ligands. The results indicated that ML models trained with a dataset exclusively composed of peptides/peptidomimetics provide limited predictive power for small molecules, but that ML models trained with a diverse dataset composed of an array of both peptides/peptidomimetics and small molecules displayed exceptional results in terms of accuracy and false rates. The diversified models can accurately differentiate the binding small molecules from non-binding small molecules using an external validation set with new small molecules that we synthesized previously. Structural features that are the most critical contributors to binding activity were extracted and are remarkably consistent with the crystallography and mutagenesis studies.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Chemistry, Lakehead University and Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON, P7B 6V4, Canada
| | - Austin M Hopkins
- Department of Chemistry, Lakehead University and Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON, P7B 6V4, Canada
| | - Peizhi Yan
- Department of Electrical and Computer Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Shan Du
- Department of Computer Science, Mathematics, Physics and Statistics, The University of British Columbia, Okanagan, Kelowna, BC, Canada
| | - Leonard G Luyt
- Department of Chemistry, University of Western Ontario, London, ON, Canada
- London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada
| | - Yifeng Li
- Department of Computer Science, Brock University, Saint Catharines, ON, Canada
| | - Jinqiang Hou
- Department of Chemistry, Lakehead University and Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON, P7B 6V4, Canada.
| |
Collapse
|
14
|
Provasi D, Filizola M. Enhancing Opioid Bioactivity Predictions through Integration of Ligand-Based and Structure-Based Drug Discovery Strategies with Transfer and Deep Learning Techniques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.04.552065. [PMID: 37609329 PMCID: PMC10441297 DOI: 10.1101/2023.08.04.552065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The opioid epidemic has cast a shadow over public health, necessitating immediate action to address its devastating consequences. To effectively combat this crisis, it is crucial to discover better opioid drugs with reduced addiction potential. Artificial intelligence-based and other machine learning tools, particularly deep learning models, have garnered significant attention in recent years for their potential to advance drug discovery. However, utilizing these tools poses challenges, especially when training samples are insufficient to achieve adequate prediction performance. In this study, we investigate the effectiveness of transfer learning using combined ligand-based and structure-based molecular descriptors from the entire opioid receptor (OR) subfamily in building robust deep learning models for enhanced bioactivity prediction of opioid ligands at each individual OR subtype. Our studies hold the potential to greatly advance opioid research by enabling the rapid identification of novel chemical probes with specific bioactivities, which can aid in the study of receptor function and contribute to the future development of improved opioid therapeutics.
Collapse
|
15
|
Wang Y, Shao L, Kang X, Zhang H, Lü F, He P. A critical review on odor measurement and prediction. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2023; 336:117651. [PMID: 36878058 DOI: 10.1016/j.jenvman.2023.117651] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/15/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
Odor pollution has become a global environmental issue of increasing concern in recent years. Odor measurements are the basis of assessing and solving odor problems. Olfactory and chemical analysis can be used for odor and odorant measurements. Olfactory analysis reflects the subjective perception of human, and chemical analysis reveals the chemical composition of odors. As an alternative to olfactory analysis, odor prediction methods have been developed based on chemical and olfactory analysis results. The combination of olfactory and chemical analysis is the best way to control odor pollution, evaluate the performances of the technologies, and predict odor. However, there are still some limitations and obstacles for each method, their combination, and the prediction. Here, we present an overview of odor measurement and prediction. Different olfactory analysis methods (namely, the dynamic olfactometry method and the triangle odor bag method) are compared in detail, the latest revisions of the standard olfactometry methods are summarized, and the uncertainties of olfactory measurement results (i.e., the odor thresholds) are analyzed. The researches, applications, and limitations of chemical analysis and odor prediction are introduced and discussed. Finally, the development and application of odor databases and algorithms for optimizing odor measurement and prediction methods are prospected, and a preliminary framework for an odor database is proposed. This review is expected to provide insights into odor measurement and prediction.
Collapse
Affiliation(s)
- Yujing Wang
- Institute of Waste Treatment & Reclamation, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Liming Shao
- Institute of Waste Treatment & Reclamation, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, China
| | - Xinyue Kang
- Institute of Waste Treatment & Reclamation, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Hua Zhang
- Institute of Waste Treatment & Reclamation, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, China
| | - Fan Lü
- Institute of Waste Treatment & Reclamation, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, China
| | - Pinjing He
- Institute of Waste Treatment & Reclamation, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, China.
| |
Collapse
|
16
|
Satake H. Kobayashi Award 2021: Neuropeptides, receptors, and follicle development in the ascidian, Ciona intestinalis Type A: New clues to the evolution of chordate neuropeptidergic systems from biological niches. Gen Comp Endocrinol 2023; 337:114262. [PMID: 36925021 DOI: 10.1016/j.ygcen.2023.114262] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023]
Abstract
Ciona intestinalis Type A (Ciona robusta) is a cosmopolitan species belonging to the phylum Urochordata, invertebrate chordates that are phylogenetically the most closely related to the vertebrates. Therefore, this species is of interest for investigation of the evolution and comparative physiology of endocrine, neuroendocrine, and nervous systems in chordates. Our group has identified>30 Ciona neuropeptides (80% of all identified ascidian neuropeptides) primarily using peptidomic approaches combined with reference to genome sequences. These neuropeptides are classified into two groups: homologs or prototypes of vertebrate neuropeptides and novel (Ciona-specific) neuropeptides. We have also identified the cognate receptors for these peptides. In particular, we elucidated multiple receptors for Ciona-specific neuropeptides by a combination of a novel machine learning system and experimental validation of the specific interaction of the predicted neuropeptide-receptor pairs, and verified unprecedented phylogenies of receptors for neuropeptides. Moreover, several neuropeptides were found to play major roles in the regulation of ovarian follicle development. Ciona tachykinin facilitates the growth of vitellogenic follicles via up-regulation of the enzymatic activities of proteases. Ciona vasopressin stimulates oocyte maturation and ovulation via up-regulation of maturation-promoting factor- and matrix metalloproteinase-directed collagen degradation, respectively. Ciona cholecystokinin also triggers ovulation via up-regulation of receptor tyrosine kinase signaling and the subsequent activation of matrix metalloproteinase. These studies revealed that the neuropeptidergic system plays major roles in ovarian follicle growth, maturation, and ovulation in Ciona, thus paving the way for investigation of the biological roles for neuropeptides in the endocrine, neuroendocrine, nervous systems of Ciona, and studies of the evolutionary processes of various neuropeptidergic systems in chordates.
Collapse
Affiliation(s)
- Honoo Satake
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan.
| |
Collapse
|
17
|
Remington JM, McKay KT, Beckage NB, Ferrell JB, Schneebeli ST, Li J. GPCRLigNet: rapid screening for GPCR active ligands using machine learning. J Comput Aided Mol Des 2023; 37:147-156. [PMID: 36840893 PMCID: PMC10379640 DOI: 10.1007/s10822-023-00497-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/03/2023] [Indexed: 02/26/2023]
Abstract
Molecules with bioactivity towards G protein-coupled receptors represent a subset of the vast space of small drug-like molecules. Here, we compare machine learning models, including dilated graph convolutional networks, that conduct binary classification to quickly identify molecules with activity towards G protein-coupled receptors. The models are trained and validated using a large set of over 600,000 active, inactive, and decoy compounds. The best performing machine learning model, dubbed GPCRLigNet, was a surprisingly simple feedforward dense neural network mapping from Morgan fingerprints to activity. Incorporation of GPCRLigNet into a high-throughput virtual screening workflow is demonstrated with molecular docking towards a particular G protein-coupled receptor, the pituitary adenylate cyclase-activating polypeptide receptor type 1. Through rigorous comparison of docking scores for molecules selected with and without using GPCRLigNet, we demonstrate an enrichment of potentially potent molecules using GPCRLigNet. This work provides a proof of principle that GPCRLigNet can effectively hone the chemical search space towards ligands with G protein-coupled receptor activity.
Collapse
Affiliation(s)
- Jacob M Remington
- Department of Chemistry, University of Vermont, Burlington, VT, 05405, USA
| | - Kyle T McKay
- Department of Chemistry, University of Vermont, Burlington, VT, 05405, USA
| | - Noah B Beckage
- Department of Chemistry, University of Vermont, Burlington, VT, 05405, USA
| | - Jonathon B Ferrell
- Department of Chemistry, University of Vermont, Burlington, VT, 05405, USA
| | - Severin T Schneebeli
- Department of Chemistry, University of Vermont, Burlington, VT, 05405, USA.,Department of Industrial and Physical Pharmacy, Department of Chemistry, Purdue University, West Lafayette, IN, 47906, USA.,Department of Pathology, University of Vermont, Burlington, VT, 05405, USA
| | - Jianing Li
- Department of Chemistry, University of Vermont, Burlington, VT, 05405, USA. .,Department of Pathology, University of Vermont, Burlington, VT, 05405, USA. .,Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47906, USA.
| |
Collapse
|
18
|
Kour S, Biswas I, Sheoran S, Arora S, Sheela P, Duppala SK, Murthy DK, Pawar SC, Singh H, Kumar D, Prabhu D, Vuree S, Kumar R. Artificial intelligence and nanotechnology for cervical cancer treatment: Current status and future perspectives. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
19
|
Durairaj J, de Ridder D, van Dijk AD. Beyond sequence: Structure-based machine learning. Comput Struct Biotechnol J 2022; 21:630-643. [PMID: 36659927 PMCID: PMC9826903 DOI: 10.1016/j.csbj.2022.12.039] [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: 09/26/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
Recent breakthroughs in protein structure prediction demarcate the start of a new era in structural bioinformatics. Combined with various advances in experimental structure determination and the uninterrupted pace at which new structures are published, this promises an age in which protein structure information is as prevalent and ubiquitous as sequence. Machine learning in protein bioinformatics has been dominated by sequence-based methods, but this is now changing to make use of the deluge of rich structural information as input. Machine learning methods making use of structures are scattered across literature and cover a number of different applications and scopes; while some try to address questions and tasks within a single protein family, others aim to capture characteristics across all available proteins. In this review, we look at the variety of structure-based machine learning approaches, how structures can be used as input, and typical applications of these approaches in protein biology. We also discuss current challenges and opportunities in this all-important and increasingly popular field.
Collapse
Affiliation(s)
- Janani Durairaj
- Biozentrum, University of Basel, Basel, Switzerland
- Bioinformatics Group, Department of Plant Sciences, Wageningen University and Research, Wageningen, the Netherlands
| | - Dick de Ridder
- Bioinformatics Group, Department of Plant Sciences, Wageningen University and Research, Wageningen, the Netherlands
| | - Aalt D.J. van Dijk
- Bioinformatics Group, Department of Plant Sciences, Wageningen University and Research, Wageningen, the Netherlands
| |
Collapse
|
20
|
Hasanzadeh A, Hamblin MR, Kiani J, Noori H, Hardie JM, Karimi M, Shafiee H. Could artificial intelligence revolutionize the development of nanovectors for gene therapy and mRNA vaccines? NANO TODAY 2022; 47:101665. [PMID: 37034382 PMCID: PMC10081506 DOI: 10.1016/j.nantod.2022.101665] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Gene therapy enables the introduction of nucleic acids like DNA and RNA into host cells, and is expected to revolutionize the treatment of a wide range of diseases. This growth has been further accelerated by the discovery of CRISPR/Cas technology, which allows accurate genomic editing in a broad range of cells and organisms in vitro and in vivo. Despite many advances in gene delivery and the development of various viral and non-viral gene delivery vectors, the lack of highly efficient non-viral systems with low cellular toxicity remains a challenge. The application of cutting-edge technologies such as artificial intelligence (AI) has great potential to find new paradigms to solve this issue. Herein, we review AI and its major subfields including machine learning (ML), neural networks (NNs), expert systems, deep learning (DL), computer vision and robotics. We discuss the potential of AI-based models and algorithms in the design of targeted gene delivery vehicles capable of crossing extracellular and intracellular barriers by viral mimicry strategies. We finally discuss the role of AI in improving the function of CRISPR/Cas systems, developing novel nanobots, and mRNA vaccine carriers.
Collapse
Affiliation(s)
- Akbar Hasanzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Noori
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Joseph M. Hardie
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02139 USA
| | - Mahdi Karimi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran 141556559, Iran
- Applied Biotechnology Research Centre, Tehran Medical Science, Islamic Azad University, Tehran 1584743311, Iran
| | - Hadi Shafiee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02139 USA
| |
Collapse
|
21
|
KUALA: a machine learning-driven framework for kinase inhibitors repositioning. Sci Rep 2022; 12:17877. [PMID: 36284125 PMCID: PMC9595087 DOI: 10.1038/s41598-022-22324-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/12/2022] [Indexed: 01/20/2023] Open
Abstract
The family of protein kinases comprises more than 500 genes involved in numerous functions. Hence, their physiological dysfunction has paved the way toward drug discovery for cancer, cardiovascular, and inflammatory diseases. As a matter of fact, Kinase binding sites high similarity has a double role. On the one hand it is a critical issue for selectivity, on the other hand, according to poly-pharmacology, a synergistic controlled effect on more than one target could be of great pharmacological interest. Another important aspect of binding similarity is the possibility of exploit it for repositioning of drugs on targets of the same family. In this study, we propose our approach called Kinase drUgs mAchine Learning frAmework (KUALA) to automatically identify kinase active ligands by using specific sets of molecular descriptors and provide a multi-target priority score and a repurposing threshold to suggest the best repurposable and non-repurposable molecules. The comprehensive list of all kinase-ligand pairs and their scores can be found at https://github.com/molinfrimed/multi-kinases .
Collapse
|
22
|
Noonan T, Denzinger K, Talagayev V, Chen Y, Puls K, Wolf CA, Liu S, Nguyen TN, Wolber G. Mind the Gap-Deciphering GPCR Pharmacology Using 3D Pharmacophores and Artificial Intelligence. Pharmaceuticals (Basel) 2022; 15:1304. [PMID: 36355476 PMCID: PMC9695541 DOI: 10.3390/ph15111304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 01/08/2025] Open
Abstract
G protein-coupled receptors (GPCRs) are amongst the most pharmaceutically relevant and well-studied protein targets, yet unanswered questions in the field leave significant gaps in our understanding of their nuanced structure and function. Three-dimensional pharmacophore models are powerful computational tools in in silico drug discovery, presenting myriad opportunities for the integration of GPCR structural biology and cheminformatics. This review highlights success stories in the application of 3D pharmacophore modeling to de novo drug design, the discovery of biased and allosteric ligands, scaffold hopping, QSAR analysis, hit-to-lead optimization, GPCR de-orphanization, mechanistic understanding of GPCR pharmacology and the elucidation of ligand-receptor interactions. Furthermore, advances in the incorporation of dynamics and machine learning are highlighted. The review will analyze challenges in the field of GPCR drug discovery, detailing how 3D pharmacophore modeling can be used to address them. Finally, we will present opportunities afforded by 3D pharmacophore modeling in the advancement of our understanding and targeting of GPCRs.
Collapse
Affiliation(s)
- Theresa Noonan
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2-4, D-14195 Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Sun Y, Jiao Y, Shi C, Zhang Y. Deep learning-based molecular dynamics simulation for structure-based drug design against SARS-CoV-2. Comput Struct Biotechnol J 2022; 20:5014-5027. [PMID: 36091720 PMCID: PMC9448712 DOI: 10.1016/j.csbj.2022.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/03/2022] [Accepted: 09/03/2022] [Indexed: 11/26/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), has led to a global pandemic. Deep learning (DL) technology and molecular dynamics (MD) simulation are two mainstream computational approaches to investigate the geometric, chemical and structural features of protein and guide the relevant drug design. Despite a large amount of research papers focusing on drug design for SARS-COV-2 using DL architectures, it remains unclear how the binding energy of the protein-protein/ligand complex dynamically evolves which is also vital for drug development. In addition, traditional deep neural networks usually have obvious deficiencies in predicting the interaction sites as protein conformation changes. In this review, we introduce the latest progresses of the DL and DL-based MD simulation approaches in structure-based drug design (SBDD) for SARS-CoV-2 which could address the problems of protein structure and binding prediction, drug virtual screening, molecular docking and complex evolution. Furthermore, the current challenges and future directions of DL-based MD simulation for SBDD are also discussed.
Collapse
Affiliation(s)
- Yao Sun
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| | - Yanqi Jiao
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| | - Chengcheng Shi
- State Key Lab of Urban Water Resource and Environment, School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| | - Yang Zhang
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| |
Collapse
|
24
|
Margulis E, Slavutsky Y, Lang T, Behrens M, Benjamini Y, Niv MY. BitterMatch: recommendation systems for matching molecules with bitter taste receptors. J Cheminform 2022; 14:45. [PMID: 35799226 PMCID: PMC9261901 DOI: 10.1186/s13321-022-00612-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/14/2022] [Indexed: 11/10/2022] Open
Abstract
Bitterness is an aversive cue elicited by thousands of chemically diverse compounds. Bitter taste may prevent consumption of foods and jeopardize drug compliance. The G protein-coupled receptors for bitter taste, TAS2Rs, have species-dependent number of subtypes and varying expression levels in extraoral tissues. Molecular recognition by TAS2R subtypes is physiologically important, and presents a challenging case study for ligand-receptor matchmaking. Inspired by hybrid recommendation systems, we developed a new set of similarity features, and created the BitterMatch algorithm that predicts associations of ligands to receptors with ~ 80% precision at ~ 50% recall. Associations for several compounds were tested in-vitro, resulting in 80% precision and 42% recall. The encouraging performance was achieved by including receptor properties and integrating experimentally determined ligand-receptor associations with chemical ligand-to-ligand similarities. BitterMatch can predict off-targets for bitter drugs, identify novel ligands and guide flavor design. The novel features capture information regarding the molecules and their receptors, which could inform various chemoinformatic tasks. Inclusion of neighbor-informed similarities improves as experimental data mounts, and provides a generalizable framework for molecule-biotarget matching.
Collapse
Affiliation(s)
- Eitan Margulis
- The Institute of Biochemistry, Food Science and Nutrition, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yuli Slavutsky
- Department of Statistics and Data Science, Faculty of Social Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tatjana Lang
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Maik Behrens
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Yuval Benjamini
- Department of Statistics and Data Science, Faculty of Social Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Masha Y Niv
- The Institute of Biochemistry, Food Science and Nutrition, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel.
| |
Collapse
|
25
|
Priya S, Tripathi G, Singh DB, Jain P, Kumar A. Machine learning approaches and their applications in drug discovery and design. Chem Biol Drug Des 2022; 100:136-153. [PMID: 35426249 DOI: 10.1111/cbdd.14057] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/30/2022] [Accepted: 04/10/2022] [Indexed: 01/04/2023]
Abstract
This review is focused on several machine learning approaches used in chemoinformatics. Machine learning approaches provide tools and algorithms to improve drug discovery. Many physicochemical properties of drugs like toxicity, absorption, drug-drug interaction, carcinogenesis, and distribution have been effectively modeled by QSAR techniques. Machine learning is a subset of artificial intelligence, and this technique has shown tremendous potential in the field of drug discovery. Techniques discussed in this review are capable of modeling non-linear datasets, as well as big data of increasing depth and complexity. Various machine learning-based approaches are being used for drug target prediction, modeling the structure of drug target, binding site prediction, ligand-based similarity searching, de novo designing of ligands with desired properties, developing scoring functions for molecular docking, building QSAR model for biological activity prediction, and prediction of pharmacokinetic and pharmacodynamic properties of ligands. In recent years, these predictive tools and models have achieved good accuracy. By the use of more related input data, relevant parameters, and appropriate algorithms, the accuracy of these predictions can be further improved.
Collapse
Affiliation(s)
- Sonal Priya
- Department of Chemistry, T. N. B. College, TMBU, Bhagalpur, India
| | - Garima Tripathi
- Department of Chemistry, T. N. B. College, TMBU, Bhagalpur, India
| | - Dev Bukhsh Singh
- Department of Biotechnology, Siddharth University, Siddharth Nagar, India
| | - Priyanka Jain
- National Institute of Plant Genome Research, New Delhi, India
| | - Abhijeet Kumar
- Department of Chemistry, Mahatma Gandhi Central University, Motihari, India
| |
Collapse
|
26
|
Chen Y, Tian X, Fan K, Zheng Y, Tian N, Fan K. The Value of Artificial Intelligence Film Reading System Based on Deep Learning in the Diagnosis of Non-Small-Cell Lung Cancer and the Significance of Efficacy Monitoring: A Retrospective, Clinical, Nonrandomized, Controlled Study. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:2864170. [PMID: 35360550 PMCID: PMC8964156 DOI: 10.1155/2022/2864170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/26/2022] [Accepted: 01/29/2022] [Indexed: 12/24/2022]
Abstract
Objective To explore the value of artificial intelligence (AI) film reading system based on deep learning in the diagnosis of non-small-cell lung cancer (NSCLC) and the significance of curative effect monitoring. Methods We retrospectively selected 104 suspected NSCLC cases from the self-built chest CT pulmonary nodule database in our hospital, and all of them were confirmed by pathological examination. The lung CT images of the selected patients were introduced into the AI reading system of pulmonary nodules, and the recording software automatically identified the nodules, and the results were compared with the results of the original image report. The nodules detected by the AI software and film readers were evaluated by two chest experts and recorded their size and characteristics. Comparison of calculation sensitivity, false positive rate evaluation of the NSCLC software, and physician's efficiency of nodule detection whether there was a significant difference between the two groups. Results The sensitivity, specificity, accuracy, positive predictive rate, and false positive rate of NSCLC diagnosed by radiologists were 72.94% (62/85), 92.06% (58/63), 81.08% (62+58/148), 92.53% (62/67), and 7.93% (5/63), respectively. The sensitivity, specificity, accuracy, positive prediction rate, and false positive rate of AI film reading system in the diagnosis of NSCLC were 94.12% (80/85), 77.77% (49/63), 87.161% (80 + 49/148), 85.11% (80/94), and 22.22% (14/63), respectively. Compared with radiologists, the sensitivity and false positive rate of artificial intelligence film reading system in the diagnosis of NSCLC were higher (P < 0.05). The sensitivity, specificity, accuracy, positive prediction rate, and negative prediction rate of artificial intelligence film reading system in evaluating the efficacy of patients with NSCLC were 87.50% (63/72), 69.23% (9/13), 84.70% (63 + 9)/85, 94.02% (63/67), and 50% (9/18), respectively. Conclusion The AI film reading system based on deep learning has higher sensitivity for the diagnosis of NSCLC than radiologists and can be used as an auxiliary detection tool for doctors to screen for NSCLC, but its false positive rate is relatively high. Attention should be paid to identification. Meanwhile, the AI film reading system based on deep learning also has a certain guiding significance for the diagnosis and treatment monitoring of NSCLC.
Collapse
Affiliation(s)
- Yunbing Chen
- Department of Computerized Tomography, Jincheng People's Hospital (Jincheng Hospital Affiliated to Changzhi Medical College), No. 456 Wenchang East Street, Jincheng, 048026 Shanxi, China
| | - Xin Tian
- Department of Computerized Tomography, Jincheng People's Hospital (Jincheng Hospital Affiliated to Changzhi Medical College), No. 456 Wenchang East Street, Jincheng, 048026 Shanxi, China
| | - Kai Fan
- Department of Computerized Tomography, Jincheng People's Hospital (Jincheng Hospital Affiliated to Changzhi Medical College), No. 456 Wenchang East Street, Jincheng, 048026 Shanxi, China
| | - Yanni Zheng
- Department of Computerized Tomography, Jincheng People's Hospital (Jincheng Hospital Affiliated to Changzhi Medical College), No. 456 Wenchang East Street, Jincheng, 048026 Shanxi, China
| | - Nannan Tian
- Department of Computerized Tomography, Jincheng People's Hospital (Jincheng Hospital Affiliated to Changzhi Medical College), No. 456 Wenchang East Street, Jincheng, 048026 Shanxi, China
| | - Ka Fan
- Department of Computerized Tomography, Jincheng People's Hospital (Jincheng Hospital Affiliated to Changzhi Medical College), No. 456 Wenchang East Street, Jincheng, 048026 Shanxi, China
| |
Collapse
|
27
|
Martinelli DD. Generative machine learning for de novo drug discovery: A systematic review. Comput Biol Med 2022; 145:105403. [PMID: 35339849 DOI: 10.1016/j.compbiomed.2022.105403] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 02/08/2023]
Abstract
Recent research on artificial intelligence indicates that machine learning algorithms can auto-generate novel drug-like molecules. Generative models have revolutionized de novo drug discovery, rendering the explorative process more efficient. Several model frameworks and input formats have been proposed to enhance the performance of intelligent algorithms in generative molecular design. In this systematic literature review of experimental articles and reviews over the last five years, machine learning models, challenges associated with computational molecule design along with proposed solutions, and molecular encoding methods are discussed. A query-based search of the PubMed, ScienceDirect, Springer, Wiley Online Library, arXiv, MDPI, bioRxiv, and IEEE Xplore databases yielded 87 studies. Twelve additional studies were identified via citation searching. Of the articles in which machine learning was implemented, six prominent algorithms were identified: long short-term memory recurrent neural networks (LSTM-RNNs), variational autoencoders (VAEs), generative adversarial networks (GANs), adversarial autoencoders (AAEs), evolutionary algorithms, and gated recurrent unit (GRU-RNNs). Furthermore, eight central challenges were designated: homogeneity of generated molecular libraries, deficient synthesizability, limited assay data, model interpretability, incapacity for multi-property optimization, incomparability, restricted molecule size, and uncertainty in model evaluation. Molecules were encoded either as strings, which were occasionally augmented using randomization, as 2D graphs, or as 3D graphs. Statistical analysis and visualization are performed to illustrate how approaches to machine learning in de novo drug design have evolved over the past five years. Finally, future opportunities and reservations are discussed.
Collapse
|
28
|
Egyed A, Kiss DJ, Keserű GM. The Impact of the Secondary Binding Pocket on the Pharmacology of Class A GPCRs. Front Pharmacol 2022; 13:847788. [PMID: 35355719 PMCID: PMC8959758 DOI: 10.3389/fphar.2022.847788] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/01/2022] [Indexed: 12/19/2022] Open
Abstract
G-protein coupled receptors (GPCRs) are considered important therapeutic targets due to their pathophysiological significance and pharmacological relevance. Class A receptors represent the largest group of GPCRs that gives the highest number of validated drug targets. Endogenous ligands bind to the orthosteric binding pocket (OBP) embedded in the intrahelical space of the receptor. During the last 10 years, however, it has been turned out that in many receptors there is secondary binding pocket (SBP) located in the extracellular vestibule that is much less conserved. In some cases, it serves as a stable allosteric site harbouring allosteric ligands that modulate the pharmacology of orthosteric binders. In other cases it is used by bitopic compounds occupying both the OBP and SBP. In these terms, SBP binding moieties might influence the pharmacology of the bitopic ligands. Together with others, our research group showed that SBP binders contribute significantly to the affinity, selectivity, functional activity, functional selectivity and binding kinetics of bitopic ligands. Based on these observations we developed a structure-based protocol for designing bitopic compounds with desired pharmacological profile.
Collapse
Affiliation(s)
| | | | - György M. Keserű
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
29
|
Lee BD, Gitter A, Greene CS, Raschka S, Maguire F, Titus AJ, Kessler MD, Lee AJ, Chevrette MG, Stewart PA, Britto-Borges T, Cofer EM, Yu KH, Carmona JJ, Fertig EJ, Kalinin AA, Signal B, Lengerich BJ, Triche TJ, Boca SM. Ten quick tips for deep learning in biology. PLoS Comput Biol 2022; 18:e1009803. [PMID: 35324884 PMCID: PMC8946751 DOI: 10.1371/journal.pcbi.1009803] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Affiliation(s)
- Benjamin D. Lee
- In-Q-Tel Labs, Arlington, Virginia, United States of America
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
| | - Casey S. Greene
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Sebastian Raschka
- Department of Statistics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Finlay Maguire
- Faculty of Computer Science, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Alexander J. Titus
- University of New Hampshire, Manchester, New Hampshire, United States of America
- Bioeconomy.XYZ, Manchester, New Hampshire, United States of America
| | - Michael D. Kessler
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Alexandra J. Lee
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Genomics and Computational Biology Graduate Program, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Marc G. Chevrette
- Wisconsin Institute for Discovery and Department of Plant Pathology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Paul Allen Stewart
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Thiago Britto-Borges
- Section of Bioinformatics and Systems Cardiology, Klaus Tschira Institute for Integrative Computational Cardiology, University Hospital Heidelberg, Heidelberg, Germany
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology), University Hospital Heidelberg, Heidelberg, Germany
| | - Evan M. Cofer
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Graduate Program in Quantitative and Computational Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Juan Jose Carmona
- Philips Healthcare, Cambridge, Massachusetts, United States of America
| | - Elana J. Fertig
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, Department of Applied Mathematics and Statistics, Convergence Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Alexandr A. Kalinin
- Medical Big Data Group, Shenzhen Research Institute of Big Data, Shenzhen, China
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Brandon Signal
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Benjamin J. Lengerich
- Computer Science Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Timothy J. Triche
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
- Department of Pediatrics, College of Human Medicine, Michigan State University, East Lansing, Michigan, United States of America
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Simina M. Boca
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, District of Columbia, United States of America
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University Medical Center, Washington, DC, United States of America
- Cancer Prevention and Control Program, Lombardi Comprehensive Cancer Center, Washington, DC, United States of America
| |
Collapse
|
30
|
Kovachka S, Malloci G, Simsir M, Ruggerone P, Azoulay S, Mus-Veteau I. Inhibition of the drug efflux activity of Ptch1 as a promising strategy to overcome chemotherapy resistance in cancer cells. Eur J Med Chem 2022; 236:114306. [DOI: 10.1016/j.ejmech.2022.114306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 03/09/2022] [Accepted: 03/17/2022] [Indexed: 11/29/2022]
|
31
|
Wang S, Hou Y, Li X, Meng X, Zhang Y, Wang X. Practical Implementation of Artificial Intelligence-Based Deep Learning and Cloud Computing on the Application of Traditional Medicine and Western Medicine in the Diagnosis and Treatment of Rheumatoid Arthritis. Front Pharmacol 2022; 12:765435. [PMID: 35002704 PMCID: PMC8733656 DOI: 10.3389/fphar.2021.765435] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/09/2021] [Indexed: 12/23/2022] Open
Abstract
Rheumatoid arthritis (RA), an autoimmune disease of unknown etiology, is a serious threat to the health of middle-aged and elderly people. Although western medicine, traditional medicine such as traditional Chinese medicine, Tibetan medicine and other ethnic medicine have shown certain advantages in the diagnosis and treatment of RA, there are still some practical shortcomings, such as delayed diagnosis, improper treatment scheme and unclear drug mechanism. At present, the applications of artificial intelligence (AI)-based deep learning and cloud computing has aroused wide attention in the medical and health field, especially in screening potential active ingredients, targets and action pathways of single drugs or prescriptions in traditional medicine and optimizing disease diagnosis and treatment models. Integrated information and analysis of RA patients based on AI and medical big data will unquestionably benefit more RA patients worldwide. In this review, we mainly elaborated the application status and prospect of AI-assisted deep learning and cloud computation-oriented western medicine and traditional medicine on the diagnosis and treatment of RA in different stages. It can be predicted that with the help of AI, more pharmacological mechanisms of effective ethnic drugs against RA will be elucidated and more accurate solutions will be provided for the treatment and diagnosis of RA in the future.
Collapse
Affiliation(s)
- Shaohui Wang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ya Hou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuanhao Li
- Chengdu Second People's Hospital, Chengdu, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Zhang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaobo Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
32
|
Kawada T, Osugi T, Matsubara S, Sakai T, Shiraishi A, Yamamoto T, Satake H. Omics Studies for the Identification of Ascidian Peptides, Cognate Receptors, and Their Relevant Roles in Ovarian Follicular Development. Front Endocrinol (Lausanne) 2022; 13:858885. [PMID: 35321341 PMCID: PMC8936170 DOI: 10.3389/fendo.2022.858885] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/10/2022] [Indexed: 11/16/2022] Open
Abstract
Omics studies contribute to the elucidation of genomes and profiles of gene expression. In the ascidian Ciona intestinalis Type A (Ciona robusta), mass spectrometry (MS)-based peptidomic studies have detected numerous Ciona-specific (nonhomologous) neuropeptides as well as Ciona homologs of typical vertebrate neuropeptides and hypothalamic peptide hormones. Candidates for cognate G protein-coupled receptors (GPCRs) for these peptides have been found in the Ciona transcriptome by two ways. First, Ciona homologous GPCRs of vertebrate counterparts have been detected by sequence homology searches of cognate transcriptomes. Second, the transcriptome-derived GPCR candidates have been used for machine learning-based systematic prediction of interactions not only between Ciona homologous peptides and GPCRs but also between novel Ciona peptides and GPCRs. These data have ultimately led to experimental evidence for various Ciona peptide-GPCR interactions. Comparative transcriptomics between the wildtype and Ciona vasopressin (CiVP) gene-edited Ciona provide clues to the biological functions of CiVP in ovarian follicular development and whole body growth. Furthermore, the transcriptomes of follicles treated with peptides, such as Ciona tachykinin and cionin (a Ciona cholecystokinin homolog), have revealed key regulatory genes for Ciona follicle growth, maturation, and ovulation, eventually leading to the verification of essential and novel molecular mechanisms underlying these biological events. These findings indicate that omics studies, combined with artificial intelligence and single-cell technologies, pave the way for investigating in greater details the nervous, neuroendocrine, and endocrine systems of ascidians and the molecular and functional evolution and diversity of peptidergic regulatory networks throughout chordates.
Collapse
|
33
|
|
34
|
Vu O, Bender BJ, Pankewitz L, Huster D, Beck-Sickinger AG, Meiler J. The Structural Basis of Peptide Binding at Class A G Protein-Coupled Receptors. Molecules 2021; 27:molecules27010210. [PMID: 35011444 PMCID: PMC8746363 DOI: 10.3390/molecules27010210] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 12/15/2021] [Accepted: 12/18/2021] [Indexed: 11/16/2022] Open
Abstract
G protein-coupled receptors (GPCRs) represent the largest membrane protein family and a significant target class for therapeutics. Receptors from GPCRs’ largest class, class A, influence virtually every aspect of human physiology. About 45% of the members of this family endogenously bind flexible peptides or peptides segments within larger protein ligands. While many of these peptides have been structurally characterized in their solution state, the few studies of peptides in their receptor-bound state suggest that these peptides interact with a shared set of residues and undergo significant conformational changes. For the purpose of understanding binding dynamics and the development of peptidomimetic drug compounds, further studies should investigate the peptide ligands that are complexed to their cognate receptor.
Collapse
Affiliation(s)
- Oanh Vu
- Deparment of Chemistry, Vanderbilt University, Nashville, TN 37235, USA;
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA; (B.J.B.); (L.P.)
| | - Brian Joseph Bender
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA; (B.J.B.); (L.P.)
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Lisa Pankewitz
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA; (B.J.B.); (L.P.)
| | - Daniel Huster
- Institute for Medical Physics and Biophysics, Medical Department, Leipzig University, Härtelstr. 16–18, D-04107 Leipzig, Germany;
| | - Annette G. Beck-Sickinger
- Faculty of Life Sciences, Institute of Biochemistry, Leipzig University, Brüderstr. 34, D-04103 Leipzig, Germany;
| | - Jens Meiler
- Deparment of Chemistry, Vanderbilt University, Nashville, TN 37235, USA;
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA; (B.J.B.); (L.P.)
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Leipzig University Medical Center, Institute for Drug Discovery, Departments of Chemistry and Computer Science, Leipzig University, Brüderstr. 34, D-04103 Leipzig, Germany
- Correspondence:
| |
Collapse
|
35
|
Identification of Potential Drug Targets of Broad-Spectrum Inhibitors with a Michael Acceptor Moiety Using Shotgun Proteomics. Viruses 2021; 13:v13091756. [PMID: 34578337 PMCID: PMC8473112 DOI: 10.3390/v13091756] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 01/10/2023] Open
Abstract
The Michael addition reaction is a spontaneous and quick chemical reaction that is widely applied in various fields. This reaction is performed by conjugating an addition of nucleophiles with α, β-unsaturated carbonyl compounds, resulting in the bond formation of C-N, C-S, C-O, and so on. In the development of molecular materials, the Michael addition is not only used to synthesize chemical compounds but is also involved in the mechanism of drug action. Several covalent drugs that bond via Michael addition are regarded as anticarcinogens and anti-inflammatory drugs. Although drug development is mainly focused on pharmaceutical drug discovery, target-based discovery can provide a different perspective for drug usage. However, considerable time and labor are required to define a molecular target through molecular biological experiments. In this review, we systematically examine the chemical structures of current FDA-approved antiviral drugs for potential Michael addition moieties with α, β-unsaturated carbonyl groups, which may exert an unidentified broad-spectrum inhibitory mechanism to target viral or host factors. We thus propose that profiling the targets of antiviral agents, such as Michael addition products, can be achieved by employing a high-throughput LC-MS approach to comprehensively analyze the interaction between drugs and targets, and the subsequent drug responses in the cellular environment to facilitate drug repurposing and/or identify potential adverse effects, with a particular emphasis on the pros and cons of this shotgun proteomic approach.
Collapse
|
36
|
Rathore AS, Mishra S, Nikita S, Priyanka P. Bioprocess Control: Current Progress and Future Perspectives. Life (Basel) 2021; 11:life11060557. [PMID: 34199245 PMCID: PMC8231968 DOI: 10.3390/life11060557] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
Typical bioprocess comprises of different unit operations wherein a near optimal environment is required for cells to grow, divide, and synthesize the desired product. However, bioprocess control caters to unique challenges that arise due to non-linearity, variability, and complexity of biotech processes. This article presents a review of modern control strategies employed in bioprocessing. Conventional control strategies (open loop, closed loop) along with modern control schemes such as fuzzy logic, model predictive control, adaptive control and neural network-based control are illustrated, and their effectiveness is highlighted. Furthermore, it is elucidated that bioprocess control is more than just automation, and includes aspects such as system architecture, software applications, hardware, and interfaces, all of which are optimized and compiled as per demand. This needs to be accomplished while keeping process requirement, production cost, market value of product, regulatory constraints, and data acquisition requirements in our purview. This article aims to offer an overview of the current best practices in bioprocess control, monitoring, and automation.
Collapse
|
37
|
Hou T, Bian Y, McGuire T, Xie XQ. Integrated Multi-Class Classification and Prediction of GPCR Allosteric Modulators by Machine Learning Intelligence. Biomolecules 2021; 11:biom11060870. [PMID: 34208096 PMCID: PMC8230833 DOI: 10.3390/biom11060870] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/30/2021] [Accepted: 06/08/2021] [Indexed: 01/01/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) are the largest and most diverse group of cell surface receptors that respond to various extracellular signals. The allosteric modulation of GPCRs has emerged in recent years as a promising approach for developing target-selective therapies. Moreover, the discovery of new GPCR allosteric modulators can greatly benefit the further understanding of GPCR cell signaling mechanisms. It is critical but also challenging to make an accurate distinction of modulators for different GPCR groups in an efficient and effective manner. In this study, we focus on an 11-class classification task with 10 GPCR subtype classes and a random compounds class. We used a dataset containing 34,434 compounds with allosteric modulators collected from classical GPCR families A, B, and C, as well as random drug-like compounds. Six types of machine learning models, including support vector machine, naïve Bayes, decision tree, random forest, logistic regression, and multilayer perceptron, were trained using different combinations of features including molecular descriptors, Atom-pair fingerprints, MACCS fingerprints, and ECFP6 fingerprints. The performances of trained machine learning models with different feature combinations were closely investigated and discussed. To the best of our knowledge, this is the first work on the multi-class classification of GPCR allosteric modulators. We believe that the classification models developed in this study can be used as simple and accurate tools for the discovery and development of GPCR allosteric modulators.
Collapse
Affiliation(s)
- Tianling Hou
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screen (CCGS) Center and Pharmacometrics System Pharmacology Program, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.H.); (Y.B.); (T.M.)
- NIH National Center of Excellence for Computational Drug Abuse Research (CDAR), University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yuemin Bian
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screen (CCGS) Center and Pharmacometrics System Pharmacology Program, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.H.); (Y.B.); (T.M.)
- NIH National Center of Excellence for Computational Drug Abuse Research (CDAR), University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Terence McGuire
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screen (CCGS) Center and Pharmacometrics System Pharmacology Program, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.H.); (Y.B.); (T.M.)
- NIH National Center of Excellence for Computational Drug Abuse Research (CDAR), University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screen (CCGS) Center and Pharmacometrics System Pharmacology Program, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.H.); (Y.B.); (T.M.)
- Drug Discovery Institute, Departments of Computational Biology and of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Correspondence:
| |
Collapse
|
38
|
Killoran MP, Levin S, Boursier ME, Zimmerman K, Hurst R, Hall MP, Machleidt T, Kirkland TA, Friedman Ohana R. An Integrated Approach toward NanoBRET Tracers for Analysis of GPCR Ligand Engagement. Molecules 2021; 26:molecules26102857. [PMID: 34065854 PMCID: PMC8151276 DOI: 10.3390/molecules26102857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 01/22/2023] Open
Abstract
Gaining insight into the pharmacology of ligand engagement with G-protein coupled receptors (GPCRs) under biologically relevant conditions is vital to both drug discovery and basic research. NanoLuc-based bioluminescence resonance energy transfer (NanoBRET) monitoring competitive binding between fluorescent tracers and unmodified test compounds has emerged as a robust and sensitive method to quantify ligand engagement with specific GPCRs genetically fused to NanoLuc luciferase or the luminogenic HiBiT peptide. However, development of fluorescent tracers is often challenging and remains the principal bottleneck for this approach. One way to alleviate the burden of developing a specific tracer for each receptor is using promiscuous tracers, which is made possible by the intrinsic specificity of BRET. Here, we devised an integrated tracer discovery workflow that couples machine learning-guided in silico screening for scaffolds displaying promiscuous binding to GPCRs with a blend of synthetic strategies to rapidly generate multiple tracer candidates. Subsequently, these candidates were evaluated for binding in a NanoBRET ligand-engagement screen across a library of HiBiT-tagged GPCRs. Employing this workflow, we generated several promiscuous fluorescent tracers that can effectively engage multiple GPCRs, demonstrating the efficiency of this approach. We believe that this workflow has the potential to accelerate discovery of NanoBRET fluorescent tracers for GPCRs and other target classes.
Collapse
Affiliation(s)
- Michael P. Killoran
- Promega Corporation, 2800 Woods Hollow, Fitchburg, WI 53711, USA; (M.P.K.); (M.E.B.); (K.Z.); (R.H.); (M.P.H.); (T.M.)
| | - Sergiy Levin
- Promega Biosciences LLC, 277 Granada Drive, San Luis Obispo, CA 93401, USA; (S.L.); (T.A.K.)
| | - Michelle E. Boursier
- Promega Corporation, 2800 Woods Hollow, Fitchburg, WI 53711, USA; (M.P.K.); (M.E.B.); (K.Z.); (R.H.); (M.P.H.); (T.M.)
| | - Kristopher Zimmerman
- Promega Corporation, 2800 Woods Hollow, Fitchburg, WI 53711, USA; (M.P.K.); (M.E.B.); (K.Z.); (R.H.); (M.P.H.); (T.M.)
| | - Robin Hurst
- Promega Corporation, 2800 Woods Hollow, Fitchburg, WI 53711, USA; (M.P.K.); (M.E.B.); (K.Z.); (R.H.); (M.P.H.); (T.M.)
| | - Mary P. Hall
- Promega Corporation, 2800 Woods Hollow, Fitchburg, WI 53711, USA; (M.P.K.); (M.E.B.); (K.Z.); (R.H.); (M.P.H.); (T.M.)
| | - Thomas Machleidt
- Promega Corporation, 2800 Woods Hollow, Fitchburg, WI 53711, USA; (M.P.K.); (M.E.B.); (K.Z.); (R.H.); (M.P.H.); (T.M.)
| | - Thomas A. Kirkland
- Promega Biosciences LLC, 277 Granada Drive, San Luis Obispo, CA 93401, USA; (S.L.); (T.A.K.)
| | - Rachel Friedman Ohana
- Promega Corporation, 2800 Woods Hollow, Fitchburg, WI 53711, USA; (M.P.K.); (M.E.B.); (K.Z.); (R.H.); (M.P.H.); (T.M.)
- Correspondence: ; Tel.: +1-608-274-1181
| |
Collapse
|
39
|
Song YQ, Wu C, Wu KJ, Han QB, Miao XM, Ma DL, Leung CH. Ubiquitination Regulators Discovered by Virtual Screening for the Treatment of Cancer. Front Cell Dev Biol 2021; 9:665646. [PMID: 34055799 PMCID: PMC8149734 DOI: 10.3389/fcell.2021.665646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/15/2021] [Indexed: 12/03/2022] Open
Abstract
The ubiquitin-proteasome system oversees cellular protein degradation in order to regulate various critical processes, such as cell cycle control and DNA repair. Ubiquitination can serve as a marker for mutation, chemical damage, transcriptional or translational errors, and heat-induced denaturation. However, aberrant ubiquitination and degradation of tumor suppressor proteins may result in the growth and metastasis of cancer. Hence, targeting the ubiquitination cascade reaction has become a potential strategy for treating malignant diseases. Meanwhile, computer-aided methods have become widely accepted as fast and efficient techniques for early stage drug discovery. This review summarizes ubiquitination regulators that have been discovered via virtual screening and their applications for cancer treatment.
Collapse
Affiliation(s)
- Ying-Qi Song
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau
| | - Chun Wu
- Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Ke-Jia Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau
| | - Quan-Bin Han
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Xiang-Min Miao
- Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau
| |
Collapse
|
40
|
Jörg M, Madden KS. The right tools for the job: the central role for next generation chemical probes and chemistry-based target deconvolution methods in phenotypic drug discovery. RSC Med Chem 2021; 12:646-665. [PMID: 34124668 PMCID: PMC8152813 DOI: 10.1039/d1md00022e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
The reconnection of the scientific community with phenotypic drug discovery has created exciting new possibilities to develop therapies for diseases with highly complex biology. It promises to revolutionise fields such as neurodegenerative disease and regenerative medicine, where the development of new drugs has consistently proved elusive. Arguably, the greatest challenge in readopting the phenotypic drug discovery approach exists in establishing a crucial chain of translatability between phenotype and benefit to patients in the clinic. This remains a key stumbling block for the field which needs to be overcome in order to fully realise the potential of phenotypic drug discovery. Excellent quality chemical probes and chemistry-based target deconvolution techniques will be a crucial part of this process. In this review, we discuss the current capabilities of chemical probes and chemistry-based target deconvolution methods and evaluate the next advances necessary in order to fully support phenotypic screening approaches in drug discovery.
Collapse
Affiliation(s)
- Manuela Jörg
- School of Natural and Environmental Sciences, Newcastle University Bedson Building Newcastle upon Tyne NE1 7RU UK
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Victoria 3052 Australia
| | - Katrina S Madden
- School of Natural and Environmental Sciences, Newcastle University Bedson Building Newcastle upon Tyne NE1 7RU UK
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Victoria 3052 Australia
| |
Collapse
|
41
|
Abstract
Molecular dynamics (MD) simulations have become increasingly useful in the modern drug development process. In this review, we give a broad overview of the current application possibilities of MD in drug discovery and pharmaceutical development. Starting from the target validation step of the drug development process, we give several examples of how MD studies can give important insights into the dynamics and function of identified drug targets such as sirtuins, RAS proteins, or intrinsically disordered proteins. The role of MD in antibody design is also reviewed. In the lead discovery and lead optimization phases, MD facilitates the evaluation of the binding energetics and kinetics of the ligand-receptor interactions, therefore guiding the choice of the best candidate molecules for further development. The importance of considering the biological lipid bilayer environment in the MD simulations of membrane proteins is also discussed, using G-protein coupled receptors and ion channels as well as the drug-metabolizing cytochrome P450 enzymes as relevant examples. Lastly, we discuss the emerging role of MD simulations in facilitating the pharmaceutical formulation development of drugs and candidate drugs. Specifically, we look at how MD can be used in studying the crystalline and amorphous solids, the stability of amorphous drug or drug-polymer formulations, and drug solubility. Moreover, since nanoparticle drug formulations are of great interest in the field of drug delivery research, different applications of nano-particle simulations are also briefly summarized using multiple recent studies as examples. In the future, the role of MD simulations in facilitating the drug development process is likely to grow substantially with the increasing computer power and advancements in the development of force fields and enhanced MD methodologies.
Collapse
|
42
|
Salmaso V, Jacobson KA. Purinergic Signaling: Impact of GPCR Structures on Rational Drug Design. ChemMedChem 2020; 15:1958-1973. [PMID: 32803849 PMCID: PMC8276773 DOI: 10.1002/cmdc.202000465] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Indexed: 12/16/2022]
Abstract
The purinergic signaling system includes membrane-bound receptors for extracellular purines and pyrimidines, and enzymes/transporters that regulate receptor activation by endogenous agonists. Receptors include: adenosine (A1 , A2A , A2B, and A3 ) and P2Y (P2Y1 , P2Y2 , P2Y4 , P2Y6 , P2Y11 , P2Y12 , P2Y13 , and P2Y14 ) receptors (all GPCRs), as well as P2X receptors (ion channels). Receptor activation, especially accompanying physiological stress or damage, creates a temporal sequence of signaling to counteract this stress and either mobilize (P2Rs) or suppress (ARs) immune responses. Thus, modulation of this large signaling family has broad potential for treating chronic diseases. Experimentally determined structures represent each of the three receptor families. We focus on selective purinergic agonists (A1 , A3 ), antagonists (A3 , P2Y14 ), and allosteric modulators (P2Y1 , A3 ). Examples of applying structure-based design, including the rational modification of known ligands, are presented for antithrombotic P2Y1 R antagonists and anti-inflammatory P2Y14 R antagonists and A3 AR agonists. A3 AR agonists are a potential, nonaddictive treatment for chronic neuropathic pain.
Collapse
Affiliation(s)
- Veronica Salmaso
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenneth A Jacobson
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
43
|
|