1
|
Samathoti P, Kumarachari RK, Bukke SPN, Rajasekhar ESK, Jaiswal AA, Eftekhari Z. The role of nanomedicine and artificial intelligence in cancer health care: individual applications and emerging integrations-a narrative review. Discov Oncol 2025; 16:697. [PMID: 40338421 PMCID: PMC12061837 DOI: 10.1007/s12672-025-02469-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
Cancer remains one of the deadliest diseases globally, significantly impacting patients' quality of life. Addressing the rising incidence of cancer deaths necessitates innovative approaches such as nanomedicine and artificial intelligence (AI). The convergence of nanomedicine and AI represents a transformative frontier in cancer healthcare, promising unprecedented advancements in diagnosis, treatment, and patient management. This narrative review explores the distinct applications of nanomedicine and AI in oncology, alongside their synergistic potential. Nanomedicine leverages nanoparticles for targeted drug delivery, enhancing therapeutic efficacy while minimizing adverse effects. Concurrently, AI algorithms facilitate early cancer detection, personalized treatment planning, and predictive analytics, thereby optimizing clinical outcomes. Emerging integrations of these technologies could transform cancer care by facilitating precise, personalized, and adaptive treatment strategies. This review synthesizes current research, highlights innovative individual applications, and discusses the emerging integrations of nanomedicine and AI in oncology. The goal is to provide a comprehensive understanding of how these cutting-edge technologies can collaboratively improve cancer diagnosis, treatment, and patient prognosis.
Collapse
Affiliation(s)
- Prasanthi Samathoti
- Department of Pharmaceutics, MB School of Pharmaceutical Sciences (Earst While Sree Vidyanikethan College of Pharmacy), Mohan Babu University, Tirupati, 517102, Andhra Pradesh, India
| | - Rajasekhar Komarla Kumarachari
- Department of Pharmaceutical Chemistry, Meenakshi Faculty of Pharmacy, MAHER University, Thandalam, MevalurKuppam, 602105, Tamil Nadu, India
| | - Sarad Pawar Naik Bukke
- Department of Pharmaceutics and Pharmaceutical Technology, Kampala International University, Western Campus, P.O. Box 71, Ishaka, Bushenyi, Uganda.
| | - Eashwar Sai Komarla Rajasekhar
- Department of Data Science and Artificial Intelligence, Indian Institute of Technology, Bhilai, Kutela Bhata, 491001, Chattisgarh, India
| | | | - Zohre Eftekhari
- Department of Biotechnology, Pasteur Institute of Iran, District 11, Rajabi, M9RW+M55, Tehran, Tehran Province, Iran
| |
Collapse
|
2
|
Al-Amin M, Tanjin R, Karim MR, Etee JM, Siddika A, Akter N, Uddin MH, Mahmud R, Saffat T, Hossen MF, Mowlee SI, Rafa EK, Akter S. Anti-cancer activity elucidation of geissolosimine as an MDM2-p53 interaction inhibitor: An in-silico study. PLoS One 2025; 20:e0323003. [PMID: 40339040 PMCID: PMC12061181 DOI: 10.1371/journal.pone.0323003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/01/2025] [Indexed: 05/10/2025] Open
Abstract
For cancer treatment, Inhibition of murine double minute (MDM2) & p53 interaction is considered an attractive therapeutic approach. In this study, we performed an integrated virtual screening (i.e., QSAR, structural similarity, molecular docking, and molecular dynamic simulation) on the in-house building alkaloids library. Geissolosimine (i.e., an indole alkaloid) was predicted as a potential inhibitor for MDM2-p53 interaction. The predicted pIC50 value of Geissolosimine, was 7.013 M. Moreover, Geissolosimine showed 0.62% structural similarity to 'SAR405838' (i.e., a clinical trial inhibitor for MDM2-p53 interaction inhibition); and a docking score of -10.9 kcal/mol that was higher than the 'SAR405838'.100 ns molecular dynamics simulation (MDS) was performed to validate the docking result and it exhibited better binding stability to MDM2. The pharmacokinetic & drug-likeness analysis suggested that Geissolosimine had potential to be a drug-like compound. However, in vitro & in vivo assays will be required to validate this study.
Collapse
Affiliation(s)
- Md. Al-Amin
- Department of Pharmacy, Islamic University, Kushtia, Bangladesh
| | - Rehnuma Tanjin
- Department of Pharmacy, Islamic University, Kushtia, Bangladesh
| | - Md. Rasul Karim
- Department of Pharmacy, Islamic University, Kushtia, Bangladesh
| | | | - Ayesha Siddika
- Department of Pharmacy, Islamic University, Kushtia, Bangladesh
| | - Nafisa Akter
- Department of Pharmacy, Islamic University, Kushtia, Bangladesh
| | - Md. Helal Uddin
- Department of Pharmacy, Islamic University, Kushtia, Bangladesh
| | - Ratul Mahmud
- Department of Pharmacy, Islamic University, Kushtia, Bangladesh
| | - Tasfia Saffat
- Department of Pharmacy, Islamic University, Kushtia, Bangladesh
| | | | | | - Elmu Kabir Rafa
- Department of Pharmacy, Islamic University, Kushtia, Bangladesh
| | - Sumi Akter
- Department of Pharmacy, Islamic University, Kushtia, Bangladesh
| |
Collapse
|
3
|
Bryan JS, Pessoa P, Tavakoli M, Pressé S. Perspectives: Comparison of deep learning segmentation models on biophysical and biomedical data. Biophys J 2025; 124:1327-1339. [PMID: 40158204 DOI: 10.1016/j.bpj.2025.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 02/11/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
Deep learning-based approaches are now widely used across biophysics to help automate a variety of tasks including image segmentation, feature selection, and deconvolution. However, the presence of multiple competing deep learning architectures, each with its own advantages and disadvantages, makes it challenging to select an architecture best suited for a specific application. As such, we present a comprehensive comparison of common models. Here, we focus on the task of segmentation assuming typical (often small) training dataset sizes available from biophysics experiments and compare the following four commonly used architectures: convolutional neural networks, U-Nets, vision transformers, and vision state space models. In doing so, we establish criteria for determining optimal conditions under which each model excels, thereby offering practical guidelines for researchers and practitioners in the field.
Collapse
Affiliation(s)
- J Shepard Bryan
- Department of Physics, Arizona State University, Tempe, Arizona; Center for Biological Physics, Arizona State University, Tempe, Arizona
| | - Pedro Pessoa
- Department of Physics, Arizona State University, Tempe, Arizona; Center for Biological Physics, Arizona State University, Tempe, Arizona
| | - Meysam Tavakoli
- Department of Radiation Oncology, Emory School of Medicine, Atlanta, Georgia.
| | - Steve Pressé
- Department of Physics, Arizona State University, Tempe, Arizona; Center for Biological Physics, Arizona State University, Tempe, Arizona; School of Molecular Sciences, Arizona State University, Tempe, Arizona.
| |
Collapse
|
4
|
Cruz‐Cortés C, Fernández‐de Gortari E, Aguayo‐Ortiz R, Šeflová J, Ard A, Clasby M, Anumonwo J, Michel Espinoza‐Fonseca L. Machine Learning-Driven Discovery of Structurally Related Natural Products as Activators of the Cardiac Calcium Pump SERCA2a. ChemMedChem 2025; 20:e202400913. [PMID: 39853697 PMCID: PMC12058239 DOI: 10.1002/cmdc.202400913] [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: 11/12/2024] [Revised: 01/18/2025] [Accepted: 01/23/2025] [Indexed: 01/26/2025]
Abstract
A key molecular dysfunction in heart failure is the reduced activity of the cardiac sarcoplasmic reticulum Ca2+-ATPase (SERCA2a) in cardiac muscle cells. Reactivating SERCA2a improves cardiac function in heart failure models, making it a validated target and an attractive therapeutic approach for heart failure therapy. However, finding small-molecule SERCA2a activators is challenging. In this study, we used a machine learning-based virtual screening to identify SERCA2a activators among 57,423 natural products. The machine learning model identified ten structurally related natural products from Zingiber officinale, Aframomum melegueta, Alpinia officinarum, Alpinia oxyphylla, and Capsicum (chili peppers) as SERCA2a activators. Initial ATPase assays showed seven of these activate SERCA at low micromolar concentrations. Notably, two natural products, Yakuchinone A and Alpinoid D displayed robust concentration-dependent responses in primary ATPase activity assays, efficient lipid bilayer binding and permeation in atomistic simulations, and enhanced intracellular Ca2+ transport in adult mouse cardiac cells. While these natural products exert off-target effects on Ca2+ signaling, these compounds offer promising avenues for the design and optimization of lead compounds. In conclusion, this study increases the array of calcium pump effectors and provides new scaffolds for the development of novel SERCA2a activators as new therapies for heart failure.
Collapse
Affiliation(s)
- Carlos Cruz‐Cortés
- Center for Arrhythmia ResearchDepartment of Internal MedicineDivision of Cardiovascular MedicineUniversity of MichiganAnn ArborMI-48109USA
| | - Eli Fernández‐de Gortari
- International Iberian Nanotechnology LaboratoryBraga4715-330Portugal
- Euskal Oxcitas Biotek SLCalle Lutxana 11 – DCHA48008BilbaoSpain
| | - Rodrigo Aguayo‐Ortiz
- Departamento de FarmaciaFacultad de QuímicaUniversidad Nacional Autónoma de MéxicoMexico City04510Mexico
| | - Jaroslava Šeflová
- Department of Cell and Molecular PhysiologyLoyola University ChicagoMaywoodIL-60153USA
| | - Adam Ard
- College of PharmacyUniversity of MichiganAnn ArborMI-48109USA
- Vahlteich Medicinal Chemistry CoreUniversity of MichiganAnn ArborMI-48109USA
| | - Martin Clasby
- College of PharmacyUniversity of MichiganAnn ArborMI-48109USA
- Vahlteich Medicinal Chemistry CoreUniversity of MichiganAnn ArborMI-48109USA
| | - Justus Anumonwo
- Center for Arrhythmia ResearchDepartment of Internal MedicineDivision of Cardiovascular MedicineUniversity of MichiganAnn ArborMI-48109USA
| | - L. Michel Espinoza‐Fonseca
- Center for Arrhythmia ResearchDepartment of Internal MedicineDivision of Cardiovascular MedicineUniversity of MichiganAnn ArborMI-48109USA
| |
Collapse
|
5
|
Wu J, Qiu Y, Lyashenko E, Torregrosa T, Pfister EL, Ryan MJ, Mueller C, Choudhury SR. Prediction of Adeno-Associated Virus Fitness with a Protein Language-Based Machine Learning Model. Hum Gene Ther 2025; 36:823-829. [PMID: 40241334 DOI: 10.1089/hum.2024.227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
Adeno-associated virus (AAV)-based therapeutics have the potential to transform the lives of patients by delivering one-time treatments for a variety of diseases. However, a critical challenge to their widespread adoption and distribution is the high cost of goods. Reducing manufacturing costs by developing AAV capsids with improved yield, or fitness, is key to making gene therapies more affordable. AAV fitness is largely determined by the amino acid sequence of the capsid, however, engineered AAVs are rarely optimized for manufacturability. Here, we report a state-of-the art machine learning (ML) model that predicts the fitness of AAV2 capsid mutants based on the amino acid sequence of the capsid monomer. By combining a protein language model (PLM) and classical ML techniques, our model achieved a significantly high prediction accuracy (Pearson correlation = 0.818) for capsid fitness. Importantly, tests on completely independent datasets showed robustness and generalizability of our model, even for multimutant AAV capsids. Our accurate ML-based model can be used as a surrogate for laborious in vitro experiments, thus saving time and resources, and can be deployed to increase the fitness of clinical AAV capsids to make gene therapies economically viable for patients.
Collapse
Affiliation(s)
- Jason Wu
- Genomic Medicine Unit, Sanofi, Waltham, Massachusetts, USA
| | - Yu Qiu
- Large Molecule Research, Sanofi, Cambridge, Massachusetts, USA
| | | | | | | | - Michael J Ryan
- Genomic Medicine Unit, Sanofi, Waltham, Massachusetts, USA
| | | | | |
Collapse
|
6
|
Kumar V, Sharma N, Mishra VK, Mall S, Kumar A, Dev K, Patel CN. Computational Evaluation of Phytocompounds From Selective Medicinal Plants as Potential Antidiabetic Agents. Chem Biodivers 2025:e202403368. [PMID: 40273195 DOI: 10.1002/cbdv.202403368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 04/26/2025]
Abstract
The distinguishing characteristic of diabetes mellitus (DM), chronic hyperglycemia emphasizes the need for safer, more efficient antidiabetic treatments. This study employs computational approaches to explore the therapeutic potential of phytochemicals from medicinal plants as antidiabetic drugs. Molecular docking against phosphorylated insulin receptor (IR) tyrosine kinase and human dipeptidyl peptidase IV (DPP-IV) identified eriodictyol (-7.13 kcal/mol) and petunidin (-6.61 kcal/mol) as potent inhibitors. Molecular dynamics simulations confirmed the structural stability of these complexes, with root mean square deviation values stabilizing within 2.8-4.5 Å. Binding free energy calculations using Molecular Mechanics Generalized Born Surface Area evealed strong binding affinities of eriodictyol-IR (ΔGbinding = -44.63 ± 4.05 kcal/mol), and petunidin-DPP-IV complex (ΔGbinding = -49.86 ± 6.13 kcal/mol). Additionally, pharmacokinetic assessments showed that these compounds adhered to Lipinski's rule, with no significant hepatotoxicity or cytotoxicity. These findings underscore the potential of these phytocompounds as antidiabetic candidates, warranting further in vitro and in vivo investigations.
Collapse
Affiliation(s)
- Vikas Kumar
- University Institute of Biotechnology, Chandigarh University, Mohali, India
| | - Nitin Sharma
- Department of Biotechnology, Chandigarh Group of Colleges, Mohali, India
| | - Vipin Kumar Mishra
- Chemistry Division, School of Advanced Sciences and Language, VIT Bhopal University, Bhopal, India
| | - Smita Mall
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Ashwani Kumar
- University Institute of Biotechnology, Chandigarh University, Mohali, India
| | - Kamal Dev
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, India
- Department of Pharmacology and Toxicology, Wright State University, Dayton, Ohio, USA
| | - Chirag N Patel
- Biotechnology Research Centre, Technology Innovation Institute, Abu Dhabi, UAE
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Liu B, Zhao L, Tan Y, Yao X, Liu H, Zhang Q. Discovery and Characterization of Novel Receptor-Interacting Protein Kinase 1 Inhibitors Using Deep Learning and Virtual Screening. ACS Chem Neurosci 2025; 16:1617-1630. [PMID: 40181215 DOI: 10.1021/acschemneuro.5c00180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Receptor-interacting protein kinase 1 (RIPK1) serves as a critical mediator of cell necroptosis and represents a promising therapeutic target for various human neurodegenerative diseases and inflammatory diseases. Nonetheless, the RIPK1 inhibitors currently reported are inadequate for clinical research due to suboptimal inhibitory activities or lack of selectivity. Consequently, there is a need for the discovery of novel RIPK1 kinase inhibitors. In this study, we integrated a deep learning model, specifically the fingerprint graph attention network (FP-GAT), with molecular docking-based virtual screening to identify potential RIPK1 inhibitors from a library comprising 13 million compounds. Out of 43 compounds procured, two compounds (designated as 24 and 41) demonstrated enzyme inhibition activity exceeding 50% at a concentration of 10 μM against RIPK1. The half-maximal inhibitory concentrations (IC50) for compounds 24 and 41 were determined to be 2.01 and 2.95 μM, respectively. Furthermore, these compounds exhibited protective effects in an HT-29 cell model of TSZ-induced necroptosis, with half-maximal effective concentrations (EC50) of 6.77 μM for compound 24 and 68.70 μM for compound 41. Finally, molecular dynamics simulations and binding free energy calculations were conducted to elucidate the molecular mechanism of compounds 24 and 41 binding to RIPK1. The results show that Met92, Met95, Ala155, and Asp156 are key residues for novel RIPK1 inhibitors. In summary, this work discovered two hit compounds targeting RIPK1, which can be further structurally modified to become promising lead compounds.
Collapse
Affiliation(s)
- Bo Liu
- Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR 999078, China
| | - Likun Zhao
- Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR 999078, China
| | - Yi Tan
- Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR 999078, China
| | - Xiaojun Yao
- Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR 999078, China
| | - Huanxiang Liu
- Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR 999078, China
| | - Qianqian Zhang
- Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR 999078, China
| |
Collapse
|
8
|
T FX, R S, A K FR, S B, R K, M A, S V, S P, S A, K S, M T. Phytochemical composition, anti-microbial, anti-oxidant and anti-diabetic effects of Solanum elaeagnifolium Cav. leaves: in vitro and in silico assessments. J Biomol Struct Dyn 2025; 43:3688-3714. [PMID: 38180058 DOI: 10.1080/07391102.2023.2300124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024]
Abstract
The aim of this study was to screen the chemical components of Solanum elaeagnifolium leaves and assess their therapeutic attributes with regard to their antioxidant, antibacterial, and antidiabetic activities. The antidiabetic effects were explored to determine the α-amylase and α-glucosidase inhibitory potential of the leaf extract. To identify the active antidiabetic drugs from the extracts, the GC-MS-screened molecules were docked with diabetes-related proteins using the glide module in the Schrodinger Tool. In addition, molecular dynamics (MD) simulations were performed for 100 ns to evaluate the binding stability of the docked complex using the Desmond module. The ethyl acetate had a significant total phenolic content (TPC), with a value of 79.04 ± 0.98 mg/g GAE. The ethanol extract was tested for its minimum inhibitory concentration (MIC) for its bacteriostatic properties. It suppressed the growth of B. subtilis, E. coli, P. vulgaris, R. equi and S. epidermis at a dosage of 118.75 µg/mL. Moreover, the IC50 values of the ethanol extract were determined to be 17.78 ± 2.38 in the α-amylase and and 27.90 ± 5.02 µg/mL in α-glucosidase. The in-silico investigation revealed that cyclolaudenol achieved docking scores of -7.94 kcal/mol for α-amylase. Likewise, the α-tocopherol achieved the docking scores of -7.41 kcal/mol for glycogen phosphorylase B and -7.21 kcal/mol for phosphorylase kinase. In the MD simulations, the cyclolaudenol and α-tocopherol complexes exhibited consistently stable affinities with diabetic proteins throughout the trajectory. Based on these findings, we conclude that this plant could be a good source for the development of novel antioxidant, antibacterial, and antidiabetic agents.
Collapse
Affiliation(s)
- Francis Xavier T
- Ethnopharmacological Research Unit, PG and Research Department of Botany, St. Joseph's College (Autonomous), Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Sabitha R
- Ethnopharmacological Research Unit, PG and Research Department of Botany, St. Joseph's College (Autonomous), Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Freeda Rose A K
- PG and Research Department of Botany, Holy Cross College (Autonomous), Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Balavivekananthan S
- Ethnopharmacological Research Unit, PG and Research Department of Botany, St. Joseph's College (Autonomous), Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Kariyat R
- Department of Biology, The University of Texas, Rio Grande Valley, W University Dr, Edinburg, TX, USA
| | - Ayyanar M
- PG and Research Department of Botany, A.V.V.M. Sri Pushpam College (Autonomous), Bharathidasan University, Poondi, Tamil Nadu, India
| | - Vijayakumar S
- PG and Research Department of Botany, A.V.V.M. Sri Pushpam College (Autonomous), Bharathidasan University, Poondi, Tamil Nadu, India
| | - Prabhu S
- Division of Phytochemistry and Drug Design, Department of Biosciences, Rajagiri College of Social Sciences, Cochin, Kerala, India
| | - Amalraj S
- Division of Phytochemistry and Drug Design, Department of Biosciences, Rajagiri College of Social Sciences, Cochin, Kerala, India
| | - Shine K
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Thiruvengadam M
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, Korea
| |
Collapse
|
9
|
Basnet BB, Zhou ZY, Wei B, Wang H. Advances in AI-based strategies and tools to facilitate natural product and drug development. Crit Rev Biotechnol 2025:1-32. [PMID: 40159111 DOI: 10.1080/07388551.2025.2478094] [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: 10/20/2024] [Revised: 02/11/2025] [Accepted: 02/16/2025] [Indexed: 04/02/2025]
Abstract
Natural products and their derivatives have been important for treating diseases in humans, animals, and plants. However, discovering new structures from natural sources is still challenging. In recent years, artificial intelligence (AI) has greatly aided the discovery and development of natural products and drugs. AI facilitates to: connect genetic data to chemical structures or vice-versa, repurpose known natural products, predict metabolic pathways, and design and optimize metabolites biosynthesis. More recently, the emergence and improvement in neural networks such as deep learning and ensemble automated web based bioinformatics platforms have sped up the discovery process. Meanwhile, AI also improves the identification and structure elucidation of unknown compounds from raw data like mass spectrometry and nuclear magnetic resonance. This article reviews these AI-driven methods and tools, highlighting their practical applications and guide for efficient natural product discovery and drug development.
Collapse
Affiliation(s)
- Buddha Bahadur Basnet
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
- Central Department of Biotechnology, Tribhuvan University, Kathmandu, Nepal
| | - Zhen-Yi Zhou
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Bin Wei
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Hong Wang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Marine Fishery Resources Exploitment, Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
10
|
Jia X, Liu M, Tang Y, Meng J, Fang R, Wang X, Li C. Artificial intelligence accelerates the identification of nature-derived potent LOXL2 inhibitors. Sci Rep 2025; 15:10540. [PMID: 40148559 PMCID: PMC11950171 DOI: 10.1038/s41598-025-95530-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/21/2025] [Indexed: 03/29/2025] Open
Abstract
The role of LOXL2 in cancer has been widely demonstrated, but current therapies targeting LOXL2 are not yet fully developed. We believe that selective nature-derived inhibition of LOXL2 may provide a better therapeutic approach for the treatment of cancer. Therefore, we adopted a comprehensive approach combining deep learning and traditional computer-aided drug design methods to screen LOXL2 selective inhibitors. Bioactivity and affinity of the potential LOXL2 inhibitors were determined by molecular docking and virtual screening. At the same time, we experimentally tested the effect of potential LOXL2 inhibitors on cancer cells. Validation showed that it could inhibit proliferation and migration, promote apoptosis of CT26 cells, and reduce the expression level of LOXL2 protein. As a result, we identified a potent LOXL2 inhibitor: the natural product Forsythoside A, and demonstrated that Forsythoside A has an inhibitory effect on tumors.
Collapse
Affiliation(s)
- Xiaowei Jia
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Meng Liu
- Sijiqing Hospital, Beijing, China
| | - Yushi Tang
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jingyan Meng
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruolin Fang
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiting Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 Bei San Huan Dong Lu, Beijing, 100029, China.
| | - Cheng Li
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Tian Jin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, No.10 Poyang Lake Road, Tianjin, 301617, China.
| |
Collapse
|
11
|
Provasi D, Filizola M. Fine-Tuned Deep Transfer Learning Models for Large Screenings of Safer Drugs Targeting Class A GPCRs. Biochemistry 2025; 64:1328-1337. [PMID: 40056143 DOI: 10.1021/acs.biochem.4c00832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2025]
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 the 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, through either 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 pretrained a model on receptor sequences and ligand data sets 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
Affiliation(s)
- Davide Provasi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Marta Filizola
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
12
|
Tang X, Zhou C, Lu C, Meng Y, Xu J, Hu X, Tian G, Yang J. Enhancing Drug Repositioning Through Local Interactive Learning With Bilinear Attention Networks. IEEE J Biomed Health Inform 2025; 29:1644-1655. [PMID: 37988217 DOI: 10.1109/jbhi.2023.3335275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Drug repositioning has emerged as a promising strategy for identifying new therapeutic applications for existing drugs. In this study, we present DRGBCN, a novel computational method that integrates heterogeneous information through a deep bilinear attention network to infer potential drugs for specific diseases. DRGBCN involves constructing a comprehensive drug-disease network by incorporating multiple similarity networks for drugs and diseases. Firstly, we introduce a layer attention mechanism to effectively learn the embeddings of graph convolutional layers from these networks. Subsequently, a bilinear attention network is constructed to capture pairwise local interactions between drugs and diseases. This combined approach enhances the accuracy and reliability of predictions. Finally, a multi-layer perceptron module is employed to evaluate potential drugs. Through extensive experiments on three publicly available datasets, DRGBCN demonstrates better performance over baseline methods in 10-fold cross-validation, achieving an average area under the receiver operating characteristic curve (AUROC) of 0.9399. Furthermore, case studies on bladder cancer and acute lymphoblastic leukemia confirm the practical application of DRGBCN in real-world drug repositioning scenarios. Importantly, our experimental results from the drug-disease network analysis reveal the successful clustering of similar drugs within the same community, providing valuable insights into drug-disease interactions. In conclusion, DRGBCN holds significant promise for uncovering new therapeutic applications of existing drugs, thereby contributing to the advancement of precision medicine.
Collapse
|
13
|
Ali S, Shaikh S, Ahmad K, Choi I. Identification of active compounds as novel dipeptidyl peptidase-4 inhibitors through machine learning and structure-based molecular docking simulations. J Biomol Struct Dyn 2025; 43:1611-1620. [PMID: 38100571 DOI: 10.1080/07391102.2023.2292299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023]
Abstract
The enzyme dipeptidyl peptidase 4 (DPP4) is a potential therapeutic target for type 2 diabetes (T2DM). Many synthetic anti-DPP4 medications are available to treat T2DM. The need for secure and efficient medicines has been unmet due to the adverse side effects of existing DPP4 medications. The present study implemented a combined approach to machine learning and structure-based virtual screening to identify DPP4 inhibitors. Two ML models were trained based on DPP4 IC50 datasets. The ML models random forest (RF) and multilayer perceptron (MLP) neural network showed good accuracy, with the area under the curve being 0.93 and 0.91, respectively. The natural compound library was screened through ML models, and 1% (217) of compounds were selected for further screening. Structure-based virtual screening was performed along with positive control sitagliptin to obtain more specific and selective leads for DPP4. Based on binding affinity, drug-likeness properties, and interaction with DPP4, Z-614 and Z-997 compounds showed high binding affinity and specificity in the catalytic pocket of DPP4. Finally, the stability conformation of the DPP4 enzyme complex was checked by a molecular dynamics (MD) simulation. The MD simulation showed that both compounds bind better in the catalytic pocket, but the Z-614 compound altered the DPP4 native conformation. Therefore, Z-614 showed a high deviation in the backbone. This combined approach (ML and structure-based) study reported that Z-997 binds most stably to DPP4 in their catalytic pocket with a binding free energy of -70.3 kJ/mol, suggesting its therapeutic potential as a treatment option for T2DM disease.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shahid Ali
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
14
|
Di Stefano M, Piazza L, Poles C, Galati S, Granchi C, Giordano A, Campisi L, Macchia M, Poli G, Tuccinardi T. KinasePred: A Computational Tool for Small-Molecule Kinase Target Prediction. Int J Mol Sci 2025; 26:2157. [PMID: 40076779 PMCID: PMC11900317 DOI: 10.3390/ijms26052157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Protein kinases are key regulators of cellular processes and critical therapeutic targets in diseases like cancer, making them a focal point for drug discovery efforts. In this context, we developed KinasePred, a robust computational workflow that combines machine learning and explainable artificial intelligence to predict the kinase activity of small molecules while providing detailed insights into the structural features driving ligand-target interactions. Our kinase-family predictive tool demonstrated significant performance, validated through virtual screening, where it successfully identified six kinase inhibitors. Target-focused operational models were subsequently developed to refine target-specific predictions, enabling the identification of molecular determinants of kinase selectivity. This integrated framework not only accelerates the screening and identification of kinase-targeting compounds but also supports broader applications in target identification, polypharmacology studies, and off-target effect analysis, providing a versatile tool for streamlining the drug discovery process.
Collapse
Affiliation(s)
- Miriana Di Stefano
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Lisa Piazza
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Clarissa Poles
- Telethon Institute of Genetics and Medicine, 80078 Naples, Italy;
- Genomics and Experimental Medicine Program, Scuola Superiore Meridionale (SSM, School of Advanced Studies), 80078 Naples, Italy
| | - Salvatore Galati
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Carlotta Granchi
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Luca Campisi
- Flashtox srl, Via Tosco Romagnola 136, 56025 Pontedera, Italy;
| | - Marco Macchia
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| |
Collapse
|
15
|
Yadav K, Hasija Y. Integrated analysis of gene expressions and targeted mirnas for explaining crosstalk between oral and esophageal squamous cell carcinomas through an interpretable machine learning approach. Med Biol Eng Comput 2025; 63:483-495. [PMID: 39384707 DOI: 10.1007/s11517-024-03210-z] [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: 05/09/2024] [Accepted: 09/17/2024] [Indexed: 10/11/2024]
Abstract
This study explores the bidirectional relation of esophageal squamous cell carcinoma (ESCC) and oral squamous cell carcinoma (OSCC), examining shared risk factors and underlying molecular mechanisms. By employing random forest (RF) classifier, enhanced with interpretable machine learning (IML) through SHapley Additive exPlanations (SHAP), we analyzed gene expression from two GEO datasets (GSE30784 and GSE44021). The GSE30784 dataset comprises 167 OSCC samples and 45 control group, whereas the GSE44021 dataset encompasses 113 ESCC samples and 113 control group. Our analysis led to identification of 20 key genes, such as XBP1, VGLL1, and RAD1, which are significantly associated with development of ESCC and OSCC. Further investigations were conducted using tools like NetworkAnalyst 3.0, Single Cell Portal, and miRNET 2.0, which highlighted complex interactions between these genes and specific miRNA targets including hsa-mir-124-3p and hsa-mir-1-3p. Our model achieved high precision in identifying genes linked to crucial processes like programmed cell death and cancer pathways, suggesting new avenues for diagnosis and treatment. This study confirms the bidirectional relationship between OSCC and ESCC, laying groundwork for targeted therapeutic approaches. This study helps to identify shared biological pathways and genetic factors of these conditions for designing personalized medicine strategies and to improve disease management.
Collapse
Affiliation(s)
- Khushi Yadav
- Department of Biotechnology, Delhi Technological University (DTU), Delhi, 110042, India
| | - Yasha Hasija
- Department of Biotechnology, Delhi Technological University (DTU), Delhi, 110042, India.
| |
Collapse
|
16
|
Unsal V, Oner E, Yıldız R, Mert BD. Comparison of new secondgeneration H1 receptor blockers with some molecules; a study involving DFT, molecular docking, ADMET, biological target and activity. BMC Chem 2025; 19:4. [PMID: 39755645 DOI: 10.1186/s13065-024-01371-4] [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: 09/11/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025] Open
Abstract
Although the antiallergic properties of compounds such as CAPE, Melatonin, Curcumin, and Vitamin C have been poorly discussed by experimental studies, the antiallergic properties of these famous molecules have never been discussed with calculations. The histamine-1 receptor (H1R) belongs to the family of rhodopsin-like G-protein-coupled receptors expressed in cells that mediate allergies and other pathophysiological diseases. In this study, pharmacological activities of FDA-approved second generation H1 antihistamines (Levocetirizine, desloratadine and fexofenadine) and molecules such as CAPE, Melatonin, Curcumin, Vitamin C, ADMET (Absorption, Distribution, Metabolism, Excretion, Toxicity) profiles, density functional theory (DFT), molecular docking, biological targets and activities were compared by calculating. Since drug development is an extremely risky, costly and time-consuming process, the data obtained in this study will facilitate and guide future studies. It will also enable researchers to focus on the most promising compounds, providing an effective design strategy. Their pharmacological activity was carried out using computer-based computational techniques including DFT, molecular docking, ADMET analysis, biological targeting, and activity methods. The best binding sites of Desloratadine, Levocetirizine, Fexofenadine, CAPE, Quercetin, Melatonin, curcumin, Vitamin C ligands to Desmoglein 1, Human Histamine H1 receptor, IgE and IL13 protons were determined by molecular docking method and binding energy and interaction states were analyzed. Fexofenadine and Quercetin ligand showed the most effective binding affinity. Melatonin had the best Caco-2 permeability PPB values of Quercetin, CAPE and Curcumin were at optimal levels. On the OATP1B1 and OATP1B3 of curcumin and CAPE, Quercetin was found to have strong inhibition effects on BCRP. Melatonin and CAPE were found to have the highest inhibition values on CYP1A2, while CAPE had the highest inhibition values on CYP2C19 and CYP2C9. Vitamin C and Quercetin were found to be safer in terms of cardiac toxicity and mutagenic risks, while Desloratadine and Levocetirizine carried high risks of neurotoxicity and hematotoxicity, while CAPE was noted for its high enzyme inhibitory activities and low toxicity profiles, while the hERG blockade, DILI, and cytotoxicity values of other compounds pointed to various safety concerns. This study demonstrated the potential of machine learning methods in understanding and discovering H1 receptor blockers. The results obtained provide important clues in the development of important strategies in the clinical use of H1 receptor blockers. In the light of these data, CAPE and Quercetin are remarkable molecules.
Collapse
Affiliation(s)
- Velid Unsal
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Mardin Artuklu University, 47100, Mardin, Türkiye.
| | - Erkan Oner
- Department of Biochemistry, Faculty of Pharmacy, Adıyaman University, 02000, Adıyaman, Türkiye
| | - Reşit Yıldız
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Mardin Artuklu University, 47100, Mardin, Türkiye
| | - Başak Doğru Mert
- Energy Systems Engineering Department, Engineering Faculty, Adana Alparslan Türkeş Science and Technology University, 01250, Adana, Türkiye
| |
Collapse
|
17
|
Li R, Wu T, Xu X, Duan X, Wang Y. Deep learning-based discovery of compounds for blood pressure lowering effects. Sci Rep 2025; 15:54. [PMID: 39747442 PMCID: PMC11697042 DOI: 10.1038/s41598-024-83924-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
The hypotensive side effects caused by drugs during their use have been a vexing issue. Recent studies have found that deep learning can effectively predict the biological activity of compounds by mining patterns and rules in the data, providing a potential solution for identifying drug side effects. In this study, we established a deep learning-based predictive model, utilizing a data set comprised of compounds known to either elevate or lower blood pressure. Subsequently, the trained model was used to predict the blood pressure-lowering effects of 26,000 compounds. Based on the predicted results, we randomly selected 50 molecules for validation and compared them with literature reports. The results showed that the predictions for 30 molecules were consistent with literature reports, with known antihypertensive drugs such as reserpine, guanethidine, and mecamylamine ranking at the top. We further selected 10 of these molecules and 3 related protein targets for molecular docking, and the docking results indirectly confirmed the model's accuracy. Ultimately, we discovered and validated that salaprinol significantly inhibits ACE1 activity and lowers canine blood pressure. In summary, we have established a highly accurate activity prediction model and confirmed its accuracy in predicting potential blood pressure-lowering compounds, which is expected to help patients avoid hypotensive side effects during clinical medication and also provide significant assistance in the discovery of antihypertensive drugs.
Collapse
Affiliation(s)
- Rongzhen Li
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Tianchi Wu
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Xiaotian Xu
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Xiaoqun Duan
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China.
- School of Biomedical Industry, Guilin Medical University, Guilin, 541199, China.
| | - Yuhui Wang
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China.
- School of Biomedical Industry, Guilin Medical University, Guilin, 541199, China.
| |
Collapse
|
18
|
Khorramfard A, Pirgazi J, Ghanbari Sorkhi A. Predicting drug protein interactions based on improved support vector data description in unbalanced data. BIOIMPACTS : BI 2024; 15:30468. [PMID: 40256215 PMCID: PMC12008248 DOI: 10.34172/bi.30468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/24/2024] [Accepted: 09/07/2024] [Indexed: 04/22/2025]
Abstract
Introduction Predicting drug-protein interactions is critical in drug discovery, but traditional laboratory methods are expensive and time-consuming. Computational approaches, especially those leveraging machine learning, are increasingly popular. This paper introduces VASVDD, a multi-step method to predict drug-protein interactions. First, it extracts features from amino acid sequences in proteins and drug structures. To address the challenge of unbalanced datasets, a Support Vector Data Description (SVDD) approach is employed, outperforming standard techniques like SMOTE and ENN in balancing data. Subsequently, dimensionality reduction using a Variational Autoencoder (VAE) reduces features from 1074 to 32, improving computational efficiency and predictive performance. Methods The proposed method was evaluated on four datasets related to enzymes, G-protein-coupled receptors, ion channels, and nuclear receptors. Without preprocessing, the Gradient Boosting Classifier showed bias towards the majority class. However, balancing and dimensionality reduction significantly improved accuracy, sensitivity, specificity, and F1 scores. VASVDD demonstrated superior performance compared to other dimensionality reduction methods, such as kernel principal component analysis (kernel PCA) and Principal Component Analysis (PCA), and was validated across multiple classifiers, achieving higher AUROC values than existing techniques. Results The results highlight VASVDD's effectiveness and generalizability in predicting drug-target interactions. The method outperforms state-of-the-art techniques in terms of accuracy, robustness, and efficiency, making it a promising tool in bioinformatics for drug discovery. Conclusion The datasets analyzed during the current study are not publicly available but are available from the corresponding author upon reasonable request and source code are available on GitHub: https://github.com/alirezakhorramfard/vasvdd.
Collapse
Affiliation(s)
- Alireza Khorramfard
- Department of Electrical and Computer Engineering, University of Science and Technology of Mazandaran, Behshahr, Iran
| | - Jamshid Pirgazi
- Department of Electrical and Computer Engineering, University of Science and Technology of Mazandaran, Behshahr, Iran
| | - Ali Ghanbari Sorkhi
- Department of Electrical and Computer Engineering, University of Science and Technology of Mazandaran, Behshahr, Iran
| |
Collapse
|
19
|
Etemadifar A, Bahraminejad S, Pardakhty A, Sharifi I, Keyhani A, Ranjbar M. Preparation, Characterization, and Leishmanicidal Assessment of Silver- and Dapsone-Loaded Niosomes Co-administration: In Silico and In Vitro Study. Adv Pharm Bull 2024; 14:892-907. [PMID: 40190665 PMCID: PMC11970491 DOI: 10.34172/apb.42740] [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: 01/26/2024] [Revised: 09/17/2024] [Accepted: 12/03/2024] [Indexed: 04/09/2025] Open
Abstract
Purpose Currently, there is a crucial need for alternative strategies to control leishmaniasis, which threatens more than 1 billion people worldwide. The simultaneous use of combination therapy and nanostructured lipid carriers aimed to assess the leishmanicidal activity of silver and dapsone niosomes co-administration in vitro and in silico. Methods After preparing the niosomal formulations of dapsone and silver using the film hydration method, Span 40 and Tween 40/cholesterol with a 7/3 molar ratio was selected as the optimal formulation. Consequently, the arrays of experimental approaches were conducted to compare the anti-leishmaniasis efficacy of ready niosomes with amphotericin B and obtain a deeper understanding of their possible mechanisms of action. Results Our findings showed higher potency of silver-loaded and dapsone-loaded niosomes co-administration compared to amphotericin B as a positive control group. The results of isobologram and combination index (CI) analyses confirmed the synergic potential of this mixture. This combination triggered anti-leishmanial pathways of macrophages, which promoted the expression level of Th1 cell-related genes, and the downregulated expression of the phenotypes related to Th2 cells. Furthermore, a high level of antioxidant and apoptotic profiles against the parasite provides a rational basis and potential drug combination for cutaneous leishmaniasis. Moreover, the outcomes of the molecular docking showed a high binding affinity between dapsone and iNOS, stimulating the immune response in Th1 direction. Conclusion In general, the multifunctional leishmanicidal activity of dapsone and silver-niosomes co-administration should be considered for further in vivo and clinical studies.
Collapse
Affiliation(s)
- Anna Etemadifar
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Sina Bahraminejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Abbas Pardakhty
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Keyhani
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Ranjbar
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
20
|
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] [Grants] [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
|
21
|
Xiang ZR, Fan SR, Ren J, Ruan T, Chen Y, Zhang YW, Wang YT, Yu ZZ, Wang CF, Sun XL, Hao XJ, Chen DZ. Utilizing artificial intelligence for precision exploration of N protein targeting phenanthridine sars-cov-2 inhibitors: A novel approach. Eur J Med Chem 2024; 279:116885. [PMID: 39307103 DOI: 10.1016/j.ejmech.2024.116885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 10/28/2024]
Abstract
The persistent mutation of the novel coronavirus presents a continual threat of infections and associated illnesses. While considerable research efforts have concentrated on the functional proteins of SARS-CoV-2 in the development of anti-COVID-19 therapeutics, the structural proteins, particularly the N protein, have received comparatively less attention. This study focuses on the N protein, a critical structural component of the virus, and employs advanced deep learning models, including EMPIRE and DeepFrag, to optimize the structures of phenanthridine-based compounds. More than 10,000 small molecules, derived through deep learning, underwent high-throughput virtual screening, resulting in the synthesis of 44 compounds. Compound 38 showed a binding potential energy of -8.2 kcal/mol in molecular docking. Surface Plasmon Resonance (SPR) and Microscale Thermophoresis (MST) validation yielded dissociation constants of 353 nM and 726 nM, confirming strong binding to the N protein. Compound 38 demonstrated antiviral activity in vitro and exhibited anti-COVID-19 effects by interfering with the binding of N proteins to RNA. This research underscores the potential of targeting the SARS-CoV-2 N protein for therapeutic intervention and illustrates the efficacy of deep learning model in the design of lead compounds. The application of these deep learning models represents a promising approach for accelerating the discovery and development of antiviral agents.
Collapse
Affiliation(s)
- Zheng-Rui Xiang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Shi-Rui Fan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Juan Ren
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Ting Ruan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Yuan Chen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Yun-Wu Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China
| | - Yi-Ting Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Ze-Zhou Yu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Chao-Fan Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Xiao-Long Sun
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China; Institute of International Rivers and Eco-Security, Yunnan University, Kunming, Yunnan, 650091, China
| | - Xiao-Jiang Hao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| | - Duo-Zhi Chen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| |
Collapse
|
22
|
Zhang O, Huang Y, Cheng S, Yu M, Zhang X, Lin H, Zeng Y, Wang M, Wu Z, Zhao H, Zhang Z, Hua C, Kang Y, Cui S, Pan P, Hsieh CY, Hou T. FragGen: towards 3D geometry reliable fragment-based molecular generation. Chem Sci 2024; 15:19452-19465. [PMID: 39568888 PMCID: PMC11575641 DOI: 10.1039/d4sc04620j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/11/2024] [Indexed: 11/22/2024] Open
Abstract
3D structure-based molecular generation is a successful application of generative AI in drug discovery. Most earlier models follow an atom-wise paradigm, generating molecules with good docking scores but poor molecular properties (like synthesizability and drugability). In contrast, fragment-wise generation offers a promising alternative by assembling chemically viable fragments. However, the co-design of plausible chemical and geometrical structures is still challenging, as evidenced by existing models. To address this, we introduce the Deep Geometry Handling protocol, which decomposes the entire geometry into multiple sets of geometric variables, looking beyond model architecture design. Drawing from a newly defined six-category taxonomy, we propose FragGen, a novel hybrid strategy as the first geometry-reliable, fragment-wise molecular generation method. FragGen significantly enhances both the geometric quality and synthesizability of the generated molecules, overcoming major limitations of previous models. Moreover, FragGen has been successfully applied in real-world scenarios, notably in designing type II kinase inhibitors at the ∼nM level, establishing it as the first validated 3D fragment-based drug design algorithm. We believe that this concept-algorithm-application cycle will not only inspire researchers working on other geometry-centric tasks to move beyond architecture designs but also provide a solid example of how generative AI can be customized for drug design.
Collapse
Affiliation(s)
- Odin Zhang
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Yufei Huang
- Zhejiang University Hangzhou 310058 Zhejiang China
| | - Shichen Cheng
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Mengyao Yu
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Xujun Zhang
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Haitao Lin
- Zhejiang University Hangzhou 310058 Zhejiang China
| | - Yundian Zeng
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Mingyang Wang
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Zhenxing Wu
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Huifeng Zhao
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Zaixi Zhang
- Anhui Province Key Lab of Big Data Analysis and Application, University of Science and Technology of China Hefei Anhui China
| | - Chenqing Hua
- Montreal Institute for Learning Algorithms, McGill University Montreal QC Canada
| | - Yu Kang
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Sunliang Cui
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Peichen Pan
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Chang-Yu Hsieh
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| |
Collapse
|
23
|
Qin D, Liang X, Jiao L, Wang R, Zhao Y, Xue W, Wang J, Liang G. Sequence-Activity Relationship of Angiotensin-Converting Enzyme Inhibitory Peptides Derived from Food Proteins, Based on a New Deep Learning Model. Foods 2024; 13:3550. [PMID: 39593966 PMCID: PMC11592644 DOI: 10.3390/foods13223550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Food-derived peptides are usually safe natural drug candidates that can potentially inhibit the angiotensin-converting enzyme (ACE). The wet experiments used to identify ACE inhibitory peptides (ACEiPs) are time-consuming and costly, making it important and urgent to reduce the scope of experimental validation through bioinformatics methods. Here, we construct an ACE inhibitory peptide predictor (ACEiPP) using optimized amino acid descriptors (AADs) and long- and short-term memory neural networks. Our results show that combined-AAD models exhibit more efficient feature transformation ability than single-AAD models, especially the training model with the optimal descriptors as the feature inputs, which exhibits the highest predictive ability in the independent test (Acc = 0.9479 and AUC = 0.9876), with a significant performance improvement compared to the existing three predictors. The model can effectively characterize the structure-activity relationship of ACEiPs. By combining the model with database mining, we used ACEiPP to screen four ACEiPs with multiple reported functions. We also used ACEiPP to predict peptides from 21,249 food-derived proteins in the Database of Food-derived Bioactive Peptides (DFBP) and construct a library of potential ACEiPs to facilitate the discovery of new anti-ACE peptides.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Guizhao Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; (D.Q.); (X.L.); (L.J.); (R.W.); (Y.Z.); (W.X.); (J.W.)
| |
Collapse
|
24
|
Yang R, Ma Z, Wei Z, Wang F, Yang G. Improved Ensemble Model for Insecticide Recognition by Incorporating Insect Toxicity Data. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24219-24227. [PMID: 39439124 DOI: 10.1021/acs.jafc.4c04252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Pesticide molecules, such as insecticides, play a critical role in modern agricultural production. Traditional pesticide development methods are often inefficient and expensive, while data-driven artificial intelligence (AI) techniques have emerged as a useful tool to facilitate drug discovery. However, currently available commercial pesticide data is limited, which makes the trained models unsatisfactory in terms of performance and generalization. From a domain knowledge perspective, insect toxicity data were incorporated to improve the insecticide recognition of AI models. Compared to the models trained with the original data set, the new models performed better in the external validation, and their generalization was more desirable. In addition, by integrating different types of individual models, we obtained an ensemble model with better performance. Based on this, an online platform was developed to provide researchers with free access to insecticide screening (https://dpai.ccnu.edu.cn/InsectiVS/). Finally, two potential insecticide molecules with insecticidal activity against Plutella xylostella were successfully identified in a real-world scenario. In conclusion, this idea connects the fields of AI and agricultural chemistry and is expected to have wide application in pesticide research.
Collapse
Affiliation(s)
- Ruoqi Yang
- National Pesticide Engineering Research Center, State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin 300071, P.R. China
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan 430079, P.R. China
| | - Zhepeng Ma
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan 430079, P.R. China
| | - Zhiheng Wei
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan 430079, P.R. China
| | - Fan Wang
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan 430079, P.R. China
| | - Guangfu Yang
- National Pesticide Engineering Research Center, State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin 300071, P.R. China
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan 430079, P.R. China
| |
Collapse
|
25
|
Xu C, Zheng L, Fan Q, Liu Y, Zeng C, Ning X, Liu H, Du K, Lu T, Chen Y, Zhang Y. Progress in the application of artificial intelligence in molecular generation models based on protein structure. Eur J Med Chem 2024; 277:116735. [PMID: 39098131 DOI: 10.1016/j.ejmech.2024.116735] [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: 05/20/2024] [Revised: 07/12/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
The molecular generation models based on protein structures represent a cutting-edge research direction in artificial intelligence-assisted drug discovery. This article aims to comprehensively summarize the research methods and developments by analyzing a series of novel molecular generation models predicated on protein structures. Initially, we categorize the molecular generation models based on protein structures and highlight the architectural frameworks utilized in these models. Subsequently, we detail the design and implementation of protein structure-based molecular generation models by introducing different specific examples. Lastly, we outline the current opportunities and challenges encountered in this field, intending to offer guidance and a referential framework for developing and studying new models in related fields in the future.
Collapse
Affiliation(s)
- Chengcheng Xu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Lidan Zheng
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Qing Fan
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Yingxu Liu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Chen Zeng
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Xiangzhen Ning
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Haichun Liu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Ke Du
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Tao Lu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China.
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China.
| | - Yanmin Zhang
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China.
| |
Collapse
|
26
|
Bao Z, Tom G, Cheng A, Watchorn J, Aspuru-Guzik A, Allen C. Towards the prediction of drug solubility in binary solvent mixtures at various temperatures using machine learning. J Cheminform 2024; 16:117. [PMID: 39468626 PMCID: PMC11520512 DOI: 10.1186/s13321-024-00911-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/28/2024] [Indexed: 10/30/2024] Open
Abstract
Drug solubility is an important parameter in the drug development process, yet it is often tedious and challenging to measure, especially for expensive drugs or those available in small quantities. To alleviate these challenges, machine learning (ML) has been applied to predict drug solubility as an alternative approach. However, the majority of existing ML research has focused on the predictions of aqueous solubility and/or solubility at specific temperatures, which restricts the model applicability in pharmaceutical development. To bridge this gap, we compiled a dataset of 27,000 solubility datapoints, including solubility of small molecules measured in a range of binary solvent mixtures under various temperatures. Next, a panel of ML models were trained on this dataset with their hyperparameters tuned using Bayesian optimization. The resulting top-performing models, both gradient boosted decision trees (light gradient boosting machine and extreme gradient boosting), achieved mean absolute errors (MAE) of 0.33 for LogS (S in g/100 g) on the holdout set. These models were further validated through a prospective study, wherein the solubility of four drug molecules were predicted by the models and then validated with in-house solubility experiments. This prospective study demonstrated that the models accurately predicted the solubility of solutes in specific binary solvent mixtures under different temperatures, especially for drugs whose features closely align within the solutes in the dataset (MAE < 0.5 for LogS). To support future research and facilitate advancements in the field, we have made the dataset and code openly available. Scientific contribution Our research advances the state-of-the-art in predicting solubility for small molecules by leveraging ML and a uniquely comprehensive dataset. Unlike existing ML studies that predominantly focus on solubility in aqueous solvents at fixed temperatures, our work enables prediction of drug solubility in a variety of binary solvent mixtures over a broad temperature range, providing practical insights on the modeling of solubility for realistic pharmaceutical applications. These advancements along with the open access dataset and code support significant steps in the drug development process including new molecule discovery, drug analysis and formulation.
Collapse
Affiliation(s)
- Zeqing Bao
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Gary Tom
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, ON, M5S 1M1, Canada
| | - Austin Cheng
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, ON, M5S 1M1, Canada
| | | | - Alán Aspuru-Guzik
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, ON, M5S 1M1, Canada
- Acceleration Consortium, Toronto, ON, M5S 3H6, Canada
- Lebovic Fellow, Canadian Institute for Advanced Research (CIFAR), Toronto, ON, M5S 1M1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, M5S 3E5, Canada
- Department of Materials Science and Engineering, University of Toronto, Toronto, ON, M5S 3E4, Canada
- CIFAR Artificial Intelligence Research Chair, Vector Institute, Toronto, ON, M5S 1M1, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
- Acceleration Consortium, Toronto, ON, M5S 3H6, Canada.
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, M5S 3E5, Canada.
| |
Collapse
|
27
|
Hasan R, Bhuia MS, Chowdhury R, Khan MA, Mazumder M, Yana NT, Alencar MVOBD, Ansari SA, Ansari IA, Islam MT. Piperine exerts anti-inflammatory effects and antagonises the properties of celecoxib and ketoprofen: in vivo and molecular docking studies. Nat Prod Res 2024:1-16. [PMID: 39390887 DOI: 10.1080/14786419.2024.2413039] [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: 04/22/2024] [Revised: 09/06/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
This study evaluates the anti-inflammatory effects of a natural product, piperine (PPN), using in vivo and in silico methodologies. In the in vivo segment, inflammation was induced in the right hind paw of young chicks via a formalin (50 μL) injection. PPN was orally administered at doses of 25 and 50 mg/kg with or without celecoxib (CXB) and/or ketoprofen (KPN) (42 mg/kg). The vehicle acted as the negative control group (NC). The in silico analysis predicted the drug-likeness, pharmacokinetics, and toxicity profile of PPN, along with evaluating its binding affinity and ligand-receptor interactions. Results indicate that PPN significantly (p < 0.05) reduced licking frequency and paw edoema in a dose-dependent manner. However, in combination therapy, PPN diminished the effects of both CXB and KPN. PPN showed high affinity (-8.6 kcal/mol) towards the COX-2 enzyme. Therefore, PPN exerts anti-inflammatory effects in chicks through COX-2 inhibition pathways and antagonises CXB and KPN activities.
Collapse
Affiliation(s)
- Rubel Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd, Gopalganj, Bangladesh
| | - Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd, Gopalganj, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Muhammad Ali Khan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Modhurima Mazumder
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Noshin Tasnim Yana
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | | | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Irfan Aamer Ansari
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, Bangladesh
| |
Collapse
|
28
|
Malusare A, Aggarwal V. Improving Molecule Generation and Drug Discovery with a Knowledge-enhanced Generative Model. ARXIV 2024:arXiv:2402.08790v2. [PMID: 38410649 PMCID: PMC10896363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Recent advancements in generative models have established state-of-the-art benchmarks in the generation of molecules and novel drug candidates. Despite these successes, a significant gap persists between generative models and the utilization of extensive biomedical knowledge, often systematized within knowledge graphs, whose potential to inform and enhance generative processes has not been realized. In this paper, we present a novel approach that bridges this divide by developing a framework for knowledge-enhanced generative models called KARL. We develop a scalable methodology to extend the functionality of knowledge graphs while preserving semantic integrity, and incorporate this contextual information into a generative framework to guide a diffusion-based model. The integration of knowledge graph embeddings with our generative model furnishes a robust mechanism for producing novel drug candidates possessing specific characteristics while ensuring validity and synthesizability. KARL outperforms state-of-the-art generative models on both unconditional and targeted generation tasks.
Collapse
Affiliation(s)
- Aditya Malusare
- Edwardson School of Industrial Engineering and the Institute of Cancer Research, Purdue University
| | - Vaneet Aggarwal
- Edwardson School of Industrial Engineering and the Institute of Cancer Research, Purdue University
| |
Collapse
|
29
|
Gao C, Bao W, Wang S, Zheng J, Wang L, Ren Y, Jiao L, Wang J, Wang X. DockingGA: enhancing targeted molecule generation using transformer neural network and genetic algorithm with docking simulation. Brief Funct Genomics 2024; 23:595-606. [PMID: 38582610 DOI: 10.1093/bfgp/elae011] [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: 11/09/2023] [Revised: 02/25/2024] [Accepted: 03/13/2024] [Indexed: 04/08/2024] Open
Abstract
Generative molecular models generate novel molecules with desired properties by searching chemical space. Traditional combinatorial optimization methods, such as genetic algorithms, have demonstrated superior performance in various molecular optimization tasks. However, these methods do not utilize docking simulation to inform the design process, and heavy dependence on the quality and quantity of available data, as well as require additional structural optimization to become candidate drugs. To address this limitation, we propose a novel model named DockingGA that combines Transformer neural networks and genetic algorithms to generate molecules with better binding affinity for specific targets. In order to generate high quality molecules, we chose the Self-referencing Chemical Structure Strings to represent the molecule and optimize the binding affinity of the molecules to different targets. Compared to other baseline models, DockingGA proves to be the optimal model in all docking results for the top 1, 10 and 100 molecules, while maintaining 100% novelty. Furthermore, the distribution of physicochemical properties demonstrates the ability of DockingGA to generate molecules with favorable and appropriate properties. This innovation creates new opportunities for the application of generative models in practical drug discovery.
Collapse
Affiliation(s)
- Changnan Gao
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao 266580, China
| | - Wenjie Bao
- Guanghua School of Management, Peking University, Beijing 100091, China
| | - Shuang Wang
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao 266580, China
| | - Jianyang Zheng
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao 266580, China
| | - Lulu Wang
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao 266580, China
| | - Yongqi Ren
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao 266580, China
| | - Linfang Jiao
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao 266580, China
| | - Jianmin Wang
- The Interdisciplinary Graduate Program in Integrative Biotechnology, Yonsei University, Incheon 21983, Republic of Korea
| | - Xun Wang
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao 266580, China
- High Performance Computer Research Center, Institute of Computing Technology, CAS, Beijing 100190, China
| |
Collapse
|
30
|
Boonyarit B, Yamprasert N, Kaewnuratchadasorn P, Kinchagawat J, Prommin C, Rungrotmongkol T, Nutanong S. GraphEGFR: Multi-task and transfer learning based on molecular graph attention mechanism and fingerprints improving inhibitor bioactivity prediction for EGFR family proteins on data scarcity. J Comput Chem 2024; 45:2001-2023. [PMID: 38713612 DOI: 10.1002/jcc.27388] [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: 01/08/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 05/09/2024]
Abstract
The proteins within the human epidermal growth factor receptor (EGFR) family, members of the tyrosine kinase receptor family, play a pivotal role in the molecular mechanisms driving the development of various tumors. Tyrosine kinase inhibitors, key compounds in targeted therapy, encounter challenges in cancer treatment due to emerging drug resistance mutations. Consequently, machine learning has undergone significant evolution to address the challenges of cancer drug discovery related to EGFR family proteins. However, the application of deep learning in this area is hindered by inherent difficulties associated with small-scale data, particularly the risk of overfitting. Moreover, the design of a model architecture that facilitates learning through multi-task and transfer learning, coupled with appropriate molecular representation, poses substantial challenges. In this study, we introduce GraphEGFR, a deep learning regression model designed to enhance molecular representation and model architecture for predicting the bioactivity of inhibitors against both wild-type and mutant EGFR family proteins. GraphEGFR integrates a graph attention mechanism for molecular graphs with deep and convolutional neural networks for molecular fingerprints. We observed that GraphEGFR models employing multi-task and transfer learning strategies generally achieve predictive performance comparable to existing competitive methods. The integration of molecular graphs and fingerprints adeptly captures relationships between atoms and enables both global and local pattern recognition. We further validated potential multi-targeted inhibitors for wild-type and mutant HER1 kinases, exploring key amino acid residues through molecular dynamics simulations to understand molecular interactions. This predictive model offers a robust strategy that could significantly contribute to overcoming the challenges of developing deep learning models for drug discovery with limited data and exploring new frontiers in multi-targeted kinase drug discovery for EGFR family proteins.
Collapse
Affiliation(s)
- Bundit Boonyarit
- School of Information Science and Technology, Vidyasirimedhi Institute of Science and Technology, Rayong, Thailand
| | - Nattawin Yamprasert
- School of Information, Computer, and Communication Technology, Sirindhorn International Institute of Technology, Thammasat University, Pathum Thani, Thailand
| | | | - Jiramet Kinchagawat
- School of Information Science and Technology, Vidyasirimedhi Institute of Science and Technology, Rayong, Thailand
| | - Chanatkran Prommin
- School of Information Science and Technology, Vidyasirimedhi Institute of Science and Technology, Rayong, Thailand
| | - Thanyada Rungrotmongkol
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Sarana Nutanong
- School of Information Science and Technology, Vidyasirimedhi Institute of Science and Technology, Rayong, Thailand
| |
Collapse
|
31
|
Gangwal A, Ansari A, Ahmad I, Azad AK, Wan Sulaiman WMA. Current strategies to address data scarcity in artificial intelligence-based drug discovery: A comprehensive review. Comput Biol Med 2024; 179:108734. [PMID: 38964243 DOI: 10.1016/j.compbiomed.2024.108734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/01/2024] [Accepted: 06/08/2024] [Indexed: 07/06/2024]
Abstract
Artificial intelligence (AI) has played a vital role in computer-aided drug design (CADD). This development has been further accelerated with the increasing use of machine learning (ML), mainly deep learning (DL), and computing hardware and software advancements. As a result, initial doubts about the application of AI in drug discovery have been dispelled, leading to significant benefits in medicinal chemistry. At the same time, it is crucial to recognize that AI is still in its infancy and faces a few limitations that need to be addressed to harness its full potential in drug discovery. Some notable limitations are insufficient, unlabeled, and non-uniform data, the resemblance of some AI-generated molecules with existing molecules, unavailability of inadequate benchmarks, intellectual property rights (IPRs) related hurdles in data sharing, poor understanding of biology, focus on proxy data and ligands, lack of holistic methods to represent input (molecular structures) to prevent pre-processing of input molecules (feature engineering), etc. The major component in AI infrastructure is input data, as most of the successes of AI-driven efforts to improve drug discovery depend on the quality and quantity of data, used to train and test AI algorithms, besides a few other factors. Additionally, data-gulping DL approaches, without sufficient data, may collapse to live up to their promise. Current literature suggests a few methods, to certain extent, effectively handle low data for better output from the AI models in the context of drug discovery. These are transferring learning (TL), active learning (AL), single or one-shot learning (OSL), multi-task learning (MTL), data augmentation (DA), data synthesis (DS), etc. One different method, which enables sharing of proprietary data on a common platform (without compromising data privacy) to train ML model, is federated learning (FL). In this review, we compare and discuss these methods, their recent applications, and limitations while modeling small molecule data to get the improved output of AI methods in drug discovery. Article also sums up some other novel methods to handle inadequate data.
Collapse
Affiliation(s)
- Amit Gangwal
- Department of Natural Product Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India.
| | - Azim Ansari
- Computer Aided Drug Design Center, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Gondur, Dhule, 424002, Maharashtra, India.
| | - Abul Kalam Azad
- Faculty of Pharmacy, University College of MAIWP International, Batu Caves, 68100, Kuala Lumpur, Malaysia.
| | | |
Collapse
|
32
|
Mariani R, De Vuono MC, Businaro E, Ivaldi S, Dell'Armi T, Gallo M, Ardigò D. P.O.L.A.R. Star: A New Framework Developed and Applied by One Mid-Sized Pharmaceutical Company to Drive Digital Transformation in R&D. Pharmaceut Med 2024; 38:343-353. [PMID: 39120788 PMCID: PMC11473631 DOI: 10.1007/s40290-024-00533-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
Digital transformation has become a cornerstone of innovation in pharmaceutical research and development (R&D). Pharmaceutical companies now have an imperative to embrace transformation, including mid-sized and small-sized companies despite resource limitations that do not allow economies of scale compared with larger organizations. This article describes the journey undertaken by Chiesi to develop an efficient framework to drive digital transformation along its R&D value chain with the objective of building and refreshing a clear roadmap and relevant priorities, together with identifying and enabling new digital capabilities and skills within R&D, defining tools and processes that will guide Chiesi activities in the space up to mid-long term. This work has led so far to five main achievements, which align with the steps in the framework: a strategically aligned roadmap with key focus areas for digital transformation and a dedicated team to lead the effort; a common language for data across the R&D value chain; an internal mindset that's open to innovation and participation in key external networks and consortia; a set of quick-win use cases for the new framework; and a defined set of Key Performance Indicators (KPIs) and monitoring tools for digital transformation. The work presented here demonstrates that R&D digital transformation should represent an ongoing process to enable cross-functional collaboration and integration within complex corporate environments that face an ever-growing volume of diverse data, to efficiently support business needs, and to ensure a positive impact on patient care.
Collapse
Affiliation(s)
- Riccardo Mariani
- Chiesi Farmaceutici Spa, Largo Francesco Belloli 11/A, 43122, Parma, Italy.
| | | | - Elena Businaro
- Chiesi Farmaceutici Spa, Largo Francesco Belloli 11/A, 43122, Parma, Italy
| | - Silvia Ivaldi
- Chiesi Farmaceutici Spa, Largo Francesco Belloli 11/A, 43122, Parma, Italy
| | | | | | - Diego Ardigò
- Chiesi Farmaceutici Spa, Largo Francesco Belloli 11/A, 43122, Parma, Italy
| |
Collapse
|
33
|
Gomes AFT, de Medeiros WF, Medeiros I, Piuvezam G, da Silva-Maia JK, Bezerra IWL, Morais AHDA. In Silico Screening of Therapeutic Targets as a Tool to Optimize the Development of Drugs and Nutraceuticals in the Treatment of Diabetes mellitus: A Systematic Review. Int J Mol Sci 2024; 25:9213. [PMID: 39273161 PMCID: PMC11394750 DOI: 10.3390/ijms25179213] [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: 07/18/2024] [Revised: 08/16/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The Target-Based Virtual Screening approach is widely employed in drug development, with docking or molecular dynamics techniques commonly utilized for this purpose. This systematic review (SR) aimed to identify in silico therapeutic targets for treating Diabetes mellitus (DM) and answer the question: What therapeutic targets have been used in in silico analyses for the treatment of DM? The SR was developed following the guidelines of the Preferred Reporting Items Checklist for Systematic Review and Meta-Analysis, in accordance with the protocol registered in PROSPERO (CRD42022353808). Studies that met the PECo strategy (Problem, Exposure, Context) were included using the following databases: Medline (PubMed), Web of Science, Scopus, Embase, ScienceDirect, and Virtual Health Library. A total of 20 articles were included, which not only identified therapeutic targets in silico but also conducted in vivo analyses to validate the obtained results. The therapeutic targets most frequently indicated in in silico studies were GLUT4, DPP-IV, and PPARγ. In conclusion, a diversity of targets for the treatment of DM was verified through both in silico and in vivo reassessment. This contributes to the discovery of potential new allies for the treatment of DM.
Collapse
Affiliation(s)
- Ana Francisca T. Gomes
- Nutrition Postgraduate Program, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (A.F.T.G.); (W.F.d.M.); (J.K.d.S.-M.)
| | - Wendjilla F. de Medeiros
- Nutrition Postgraduate Program, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (A.F.T.G.); (W.F.d.M.); (J.K.d.S.-M.)
| | - Isaiane Medeiros
- Biochemistry and Molecular Biology Postgraduate Program, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil;
| | - Grasiela Piuvezam
- Public Health Postgraduate Program, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil;
| | - Juliana Kelly da Silva-Maia
- Nutrition Postgraduate Program, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (A.F.T.G.); (W.F.d.M.); (J.K.d.S.-M.)
- Department of Nutrition, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil;
| | - Ingrid Wilza L. Bezerra
- Department of Nutrition, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil;
| | - Ana Heloneida de A. Morais
- Nutrition Postgraduate Program, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (A.F.T.G.); (W.F.d.M.); (J.K.d.S.-M.)
- Biochemistry and Molecular Biology Postgraduate Program, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil;
- Department of Nutrition, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil;
| |
Collapse
|
34
|
Yucel MA, Adal E, Aktekin MB, Hepokur C, Gambacorta N, Nicolotti O, Algul O. From Deep Learning to the Discovery of Promising VEGFR-2 Inhibitors. ChemMedChem 2024; 19:e202400108. [PMID: 38726553 DOI: 10.1002/cmdc.202400108] [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: 02/05/2024] [Revised: 04/24/2024] [Indexed: 07/21/2024]
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR-2) stands as a prominent therapeutic target in oncology, playing a critical role in angiogenesis, tumor growth, and metastasis. FDA-approved VEGFR-2 inhibitors are associated with diverse side effects. Thus, finding novel and more effective inhibitors is of utmost importance. In this study, a deep learning (DL) classification model was first developed and then employed to select putative active VEGFR-2 inhibitors from an in-house chemical library including 187 druglike compounds. A pool of 18 promising candidates was shortlisted and screened against VEGFR-2 by using molecular docking. Finally, two compounds, RHE-334 and EA-11, were prioritized as promising VEGFR-2 inhibitors by employing PLATO, our target fishing and bioactivity prediction platform. Based on this rationale, we prepared RHE-334 and EA-11 and successfully tested their anti-proliferative potential against MCF-7 human breast cancer cells with IC50 values of 26.78±4.02 and 38.73±3.84 μM, respectively. Their toxicities were instead challenged against the WI-38. Interestingly, expression studies indicated that, in the presence of RHE-334, VEGFR-2 was equal to 0.52±0.03, thus comparable to imatinib equal to 0.63±0.03. In conclusion, this workflow based on theoretical and experimental approaches demonstrates effective in identifying VEGFR-2 inhibitors and can be easily adapted to other medicinal chemistry goals.
Collapse
Affiliation(s)
- Mehmet Ali Yucel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erzincan Binali Yildirim University, 24002, Erzincan, Türkiye
| | - Ercan Adal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mersin University, 33160, Mersin, Türkiye
| | - Mine Buga Aktekin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mersin University, 33160, Mersin, Türkiye
| | - Ceylan Hepokur
- Department of Biochemistry, Faculty of Pharmacy, Sivas Cumhuriyet University, 58140, Sivas, Türkiye
| | - Nicola Gambacorta
- Dipartimento di Farmacia-Scienze del Farmaco, Universita 'degli Studi di Bari "Aldo Moro", Via E. Orabona, 4, Bari I, 70125, Italy
| | - Orazio Nicolotti
- Dipartimento di Farmacia-Scienze del Farmaco, Universita 'degli Studi di Bari "Aldo Moro", Via E. Orabona, 4, Bari I, 70125, Italy
| | - Oztekin Algul
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erzincan Binali Yildirim University, 24002, Erzincan, Türkiye
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mersin University, 33160, Mersin, Türkiye
| |
Collapse
|
35
|
Rezić I, Somogyi Škoc M. Computational Methodologies in Synthesis, Preparation and Application of Antimicrobial Polymers, Biomolecules, and Nanocomposites. Polymers (Basel) 2024; 16:2320. [PMID: 39204538 PMCID: PMC11359845 DOI: 10.3390/polym16162320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/05/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
The design and optimization of antimicrobial materials (polymers, biomolecules, or nanocomposites) can be significantly advanced by computational methodologies like molecular dynamics (MD), which provide insights into the interactions and stability of the antimicrobial agents within the polymer matrix, and machine learning (ML) or design of experiment (DOE), which predicts and optimizes antimicrobial efficacy and material properties. These innovations not only enhance the efficiency of developing antimicrobial polymers but also enable the creation of materials with tailored properties to meet specific application needs, ensuring safety and longevity in their usage. Therefore, this paper will present the computational methodologies employed in the synthesis and application of antimicrobial polymers, biomolecules, and nanocomposites. By leveraging advanced computational techniques such as MD, ML, or DOE, significant advancements in the design and optimization of antimicrobial materials are achieved. A comprehensive review on recent progress, together with highlights of the most relevant methodologies' contributions to state-of-the-art materials science will be discussed, as well as future directions in the field will be foreseen. Finally, future possibilities and opportunities will be derived from the current state-of-the-art methodologies, providing perspectives on the potential evolution of polymer science and engineering of novel materials.
Collapse
Affiliation(s)
- Iva Rezić
- Department of Applied Chemistry, Faculty of Textile Technology, University of Zagreb, 10000 Zagreb, Croatia
| | - Maja Somogyi Škoc
- Department of Materials Testing, Faculty of Textile Technology, University of Zagreb, 10000 Zagreb, Croatia;
| |
Collapse
|
36
|
Jia ZC, Yang X, Wu YK, Li M, Das D, Chen MX, Wu J. The Art of Finding the Right Drug Target: Emerging Methods and Strategies. Pharmacol Rev 2024; 76:896-914. [PMID: 38866560 PMCID: PMC11334170 DOI: 10.1124/pharmrev.123.001028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
Drug targets are specific molecules in biological tissues and body fluids that interact with drugs. Drug target discovery is a key component of drug discovery and is essential for the development of new drugs in areas such as cancer therapy and precision medicine. Traditional in vitro or in vivo target discovery methods are time-consuming and labor-intensive, limiting the pace of drug discovery. With the development of modern discovery methods, the discovery and application of various emerging technologies have greatly improved the efficiency of drug discovery, shortened the cycle time, and reduced the cost. This review provides a comprehensive overview of various emerging drug target discovery strategies, including computer-assisted approaches, drug affinity response target stability, multiomics analysis, gene editing, and nonsense-mediated mRNA degradation, and discusses the effectiveness and limitations of the various approaches, as well as their application in real cases. Through the review of the aforementioned contents, a general overview of the development of novel drug targets and disease treatment strategies will be provided, and a theoretical basis will be provided for those who are engaged in pharmaceutical science research. SIGNIFICANCE STATEMENT: Target-based drug discovery has been the main approach to drug discovery in the pharmaceutical industry for the past three decades. Traditional drug target discovery methods based on in vivo or in vitro validation are time-consuming and costly, greatly limiting the development of new drugs. Therefore, the development and selection of new methods in the drug target discovery process is crucial.
Collapse
Affiliation(s)
- Zi-Chang Jia
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.)
| | - Xue Yang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.)
| | - Yi-Kun Wu
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.)
| | - Min Li
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.)
| | - Debatosh Das
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.) ;
| | - Mo-Xian Chen
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.) ;
| | - Jian Wu
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, China (Z.-C.J., X.Y., Y.-K.W., M.-X.C., J.W.); The Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee (D.D.); and State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Taian, Shandong, China (M.L.) ;
| |
Collapse
|
37
|
Zulfat M, Hakami MA, Hazazi A, Mahmood A, Khalid A, Alqurashi RS, Abdalla AN, Hu J, Wadood A, Huang X. Identification of novel NLRP3 inhibitors as therapeutic options for epilepsy by machine learning-based virtual screening, molecular docking and biomolecular simulation studies. Heliyon 2024; 10:e34410. [PMID: 39170440 PMCID: PMC11336274 DOI: 10.1016/j.heliyon.2024.e34410] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 08/23/2024] Open
Abstract
The NOD-Like Receptor Protein-3 (NLRP3) inflammasome is a key therapeutic target for the treatment of epilepsy and has been reported to regulate inflammation in several neurological diseases. In this study, a machine learning-based virtual screening strategy has investigated candidate active compounds that inhibit the NLRP3 inflammasome. As machine learning-based virtual screening has the potential to accurately predict protein-ligand binding and reduce false positives outcomes compared to traditional virtual screening. Briefly, classification models were created using Support Vector Machine (SVM), Random Forest (RF), and K-Nearest Neighbor (KNN) machine learning methods. To determine the most crucial features of a molecule's activity, feature selection was carried out. By utilizing 10-fold cross-validation, the created models were analyzed. Among the generated models, the RF model obtained the best results as compared to others. Therefore, the RF model was used as a screening tool against the large chemical databases. Molecular operating environment (MOE) and PyRx software's were applied for molecular docking. Also, using the Amber Tools program, molecular dynamics (MD) simulation of potent inhibitors was carried out. The results showed that the KNN, SVM, and RF accuracy was 0.911 %, 0.906 %, and 0.946 %, respectively. Moreover, the model has shown sensitivity of 0.82 %, 0.78 %, and 0.86 % and specificity of 0.95 %, 0.96 %, and 0.98 % respectively. By applying the model to the ZINC and South African databases, we identified 98 and 39 compounds, respectively, potentially possessing anti-NLRP3 activity. Also, a molecular docking analysis produced ten ZINC and seven South African compounds that has comparable binding affinities to the reference drug. Moreover, MD analysis of the two complexes revealed that the two compounds (ZINC000009601348 and SANC00225) form stable complexes with varying amounts of binding energy. The in-silico studies indicate that both compounds most likely display their inhibitory effect by inhibiting the NLRP3 protein.
Collapse
Affiliation(s)
- Maryam Zulfat
- Department of Biochemistry, Computational Medicinal Chemistry Laboratory, Abdul Wali Khan University, Mardan, Pakistan
| | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Al-Quwayiyah-19257, Riyadh, Saudi Arabia
| | - Ali Hazazi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Arif Mahmood
- Department of Biochemistry, Quaid-i-Azam University Islamabad, Pakistan
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, Jazan 45142, Saudi Arabia
| | - Roaya S. Alqurashi
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Ashraf N. Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Junjian Hu
- Department of Central Laboratory, SSL, Central Hospital of Dongguan City, Affiliated Dongguan Shilong People's Hospital of Guangdong Medical University, Dongguan, China
| | - Abdul Wadood
- Department of Biochemistry, Computational Medicinal Chemistry Laboratory, Abdul Wali Khan University, Mardan, Pakistan
| | - Xiaoyun Huang
- Department of Neurology, Houjie Hospital and Clinical College of Guangdong Medical University, Dongguan, China
| |
Collapse
|
38
|
Li G, Li S, Liang C, Xiao Q, Luo J. Drug repositioning based on residual attention network and free multiscale adversarial training. BMC Bioinformatics 2024; 25:261. [PMID: 39118000 PMCID: PMC11308596 DOI: 10.1186/s12859-024-05893-5] [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: 06/30/2023] [Accepted: 08/06/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Conducting traditional wet experiments to guide drug development is an expensive, time-consuming and risky process. Analyzing drug function and repositioning plays a key role in identifying new therapeutic potential of approved drugs and discovering therapeutic approaches for untreated diseases. Exploring drug-disease associations has far-reaching implications for identifying disease pathogenesis and treatment. However, reliable detection of drug-disease relationships via traditional methods is costly and slow. Therefore, investigations into computational methods for predicting drug-disease associations are currently needed. RESULTS This paper presents a novel drug-disease association prediction method, RAFGAE. First, RAFGAE integrates known associations between diseases and drugs into a bipartite network. Second, RAFGAE designs the Re_GAT framework, which includes multilayer graph attention networks (GATs) and two residual networks. The multilayer GATs are utilized for learning the node embeddings, which is achieved by aggregating information from multihop neighbors. The two residual networks are used to alleviate the deep network oversmoothing problem, and an attention mechanism is introduced to combine the node embeddings from different attention layers. Third, two graph autoencoders (GAEs) with collaborative training are constructed to simulate label propagation to predict potential associations. On this basis, free multiscale adversarial training (FMAT) is introduced. FMAT enhances node feature quality through small gradient adversarial perturbation iterations, improving the prediction performance. Finally, tenfold cross-validations on two benchmark datasets show that RAFGAE outperforms current methods. In addition, case studies have confirmed that RAFGAE can detect novel drug-disease associations. CONCLUSIONS The comprehensive experimental results validate the utility and accuracy of RAFGAE. We believe that this method may serve as an excellent predictor for identifying unobserved disease-drug associations.
Collapse
Affiliation(s)
- Guanghui Li
- School of Information Engineering, East China Jiaotong University, Nanchang, China.
| | - Shuwen Li
- School of Information Engineering, East China Jiaotong University, Nanchang, China
| | - Cheng Liang
- School of Information Science and Engineering, Shandong Normal University, Jinan, China
| | - Qiu Xiao
- College of Information Science and Engineering, Hunan Normal University, Changsha, China
| | - Jiawei Luo
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China.
| |
Collapse
|
39
|
Zhao Y, Wan Q, He X. Construction of IRAK4 inhibitor activity prediction model based on machine learning. Mol Divers 2024; 28:2289-2300. [PMID: 38970641 DOI: 10.1007/s11030-024-10926-5] [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: 05/22/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Interleukin-1 receptor-associated kinase 4 (IRAK4) is a crucial serine/threonine protein kinase that belongs to the IRAK family and plays a pivotal role in Toll-like receptor (TLR) and Interleukin-1 receptor (IL-1R) signaling pathways. Due to IRAK4's significant role in immunity, inflammation, and malignancies, it has become an intriguing target for discovering and developing potent small-molecule inhibitors. Consequently, there is a pressing need for rapid and accurate prediction of IRAK4 inhibitor activity. Leveraging a comprehensive dataset encompassing activity data for 1628 IRAK4 inhibitors, we constructed a prediction model using the LightGBM algorithm and molecular fingerprints. This model achieved an R2 of 0.829, an MAE of 0.317, and an RMSE of 0.460 in independent testing. To further validate the model's generalization ability, we tested it on 90 IRAK4 inhibitors collected in 2023. Subsequently, we applied the model to predict the activity of 13,268 compounds with docking scores less than - 9.503 kcal/mol. These compounds were initially screened from a pool of 1.6 million molecules in the chemdiv database through high-throughput molecular docking. Among these, 259 compounds with predicted pIC50 values greater than or equal to 8.00 were identified. We then performed ADMET predictions on these selected compounds. Finally, through a rigorous screening process, we identified 34 compounds that adhere to the four complementary drug-likeness rules, making them promising candidates for further investigation. Additionally, molecular dynamics simulations confirmed the stable binding of the screened compounds to the IRAK4 protein. Overall, this work presents a machine learning model for accurate prediction of IRAK4 inhibitor activity and offers new insights for subsequent structure-guided design of novel IRAK4 inhibitors.
Collapse
Affiliation(s)
- Yihuan Zhao
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, 563006, People's Republic of China.
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China.
- The Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi, 563006, China.
| | - Qianwen Wan
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, 563006, People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China
- The Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi, 563006, China
| | - Xiaoyu He
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, 563006, People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China
- The Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi, 563006, China
| |
Collapse
|
40
|
Duo L, Liu Y, Ren J, Tang B, Hirst JD. Artificial intelligence for small molecule anticancer drug discovery. Expert Opin Drug Discov 2024; 19:933-948. [PMID: 39074493 DOI: 10.1080/17460441.2024.2367014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/07/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION The transition from conventional cytotoxic chemotherapy to targeted cancer therapy with small-molecule anticancer drugs has enhanced treatment outcomes. This approach, which now dominates cancer treatment, has its advantages. Despite the regulatory approval of several targeted molecules for clinical use, challenges such as low response rates and drug resistance still persist. Conventional drug discovery methods are costly and time-consuming, necessitating more efficient approaches. The rise of artificial intelligence (AI) and access to large-scale datasets have revolutionized the field of small-molecule cancer drug discovery. Machine learning (ML), particularly deep learning (DL) techniques, enables the rapid identification and development of novel anticancer agents by analyzing vast amounts of genomic, proteomic, and imaging data to uncover hidden patterns and relationships. AREA COVERED In this review, the authors explore the important landmarks in the history of AI-driven drug discovery. They also highlight various applications in small-molecule cancer drug discovery, outline the challenges faced, and provide insights for future research. EXPERT OPINION The advent of big data has allowed AI to penetrate and enable innovations in almost every stage of medicine discovery, transforming the landscape of oncology research through the development of state-of-the-art algorithms and models. Despite challenges in data quality, model interpretability, and technical limitations, advancements promise breakthroughs in personalized and precision oncology, revolutionizing future cancer management.
Collapse
Affiliation(s)
- Lihui Duo
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Yu Liu
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Jianfeng Ren
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Bencan Tang
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Jonathan D Hirst
- School of Chemistry, University of Nottingham University Park, Nottingham, UK
| |
Collapse
|
41
|
Liu Y, Xu C, Yang X, Zhang Y, Chen Y, Liu H. Application progress of deep generative models in de novo drug design. Mol Divers 2024; 28:2411-2427. [PMID: 39097862 DOI: 10.1007/s11030-024-10942-5] [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: 05/24/2024] [Accepted: 07/16/2024] [Indexed: 08/05/2024]
Abstract
The deep molecular generative model has recently become a research hotspot in pharmacy. This paper analyzes a large number of recent reports and reviews these models. In the central part of this paper, four compound databases and two molecular representation methods are compared. Five model architectures and applications for deep molecular generative models are emphatically introduced. Three evaluation metrics for model evaluation are listed. Finally, the limitations and challenges in this field are discussed to provide a reference and basis for developing and researching new models published in future.
Collapse
Affiliation(s)
- Yingxu Liu
- School of Science, China Pharmaceutical University, Nanjing, 210009, China
| | - Chengcheng Xu
- School of Science, China Pharmaceutical University, Nanjing, 210009, China
| | - Xinyi Yang
- School of Science, China Pharmaceutical University, Nanjing, 210009, China
| | - Yanmin Zhang
- School of Science, China Pharmaceutical University, Nanjing, 210009, China
| | - Yadong Chen
- School of Science, China Pharmaceutical University, Nanjing, 210009, China
| | - Haichun Liu
- School of Science, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
42
|
Salam A, Ullah F, Amin F, Ahmad Khan I, Garcia Villena E, Kuc Castilla A, de la Torre I. Efficient prediction of anticancer peptides through deep learning. PeerJ Comput Sci 2024; 10:e2171. [PMID: 39145253 PMCID: PMC11323142 DOI: 10.7717/peerj-cs.2171] [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: 04/29/2024] [Accepted: 06/11/2024] [Indexed: 08/16/2024]
Abstract
Background Cancer remains one of the leading causes of mortality globally, with conventional chemotherapy often resulting in severe side effects and limited effectiveness. Recent advancements in bioinformatics and machine learning, particularly deep learning, offer promising new avenues for cancer treatment through the prediction and identification of anticancer peptides. Objective This study aimed to develop and evaluate a deep learning model utilizing a two-dimensional convolutional neural network (2D CNN) to enhance the prediction accuracy of anticancer peptides, addressing the complexities and limitations of current prediction methods. Methods A diverse dataset of peptide sequences with annotated anticancer activity labels was compiled from various public databases and experimental studies. The sequences were preprocessed and encoded using one-hot encoding and additional physicochemical properties. The 2D CNN model was trained and optimized using this dataset, with performance evaluated through metrics such as accuracy, precision, recall, F1-score, and area under the receiver operating characteristic curve (AUC-ROC). Results The proposed 2D CNN model achieved superior performance compared to existing methods, with an accuracy of 0.87, precision of 0.85, recall of 0.89, F1-score of 0.87, and an AUC-ROC value of 0.91. These results indicate the model's effectiveness in accurately predicting anticancer peptides and capturing intricate spatial patterns within peptide sequences. Conclusion The findings demonstrate the potential of deep learning, specifically 2D CNNs, in advancing the prediction of anticancer peptides. The proposed model significantly improves prediction accuracy, offering a valuable tool for identifying effective peptide candidates for cancer treatment. Future Work Further research should focus on expanding the dataset, exploring alternative deep learning architectures, and validating the model's predictions through experimental studies. Efforts should also aim at optimizing computational efficiency and translating these predictions into clinical applications.
Collapse
Affiliation(s)
- Abdu Salam
- Department of Computer Science, Abdul Wali Khan University, Mardan, Pakistan
| | - Faizan Ullah
- Department of Computer Science, Bacha Khan University, Charsadda, Pakistan
| | - Farhan Amin
- School of Computer Science and Engineering, Yeungnam University, Gyeongsan, Republic of Korea
| | - Izaz Ahmad Khan
- Department of Computer Science, Bacha Khan University, Charsadda, Pakistan
| | | | | | | |
Collapse
|
43
|
Vittoria Togo M, Mastrolorito F, Orfino A, Graps EA, Tondo AR, Altomare CD, Ciriaco F, Trisciuzzi D, Nicolotti O, Amoroso N. Where developmental toxicity meets explainable artificial intelligence: state-of-the-art and perspectives. Expert Opin Drug Metab Toxicol 2024; 20:561-577. [PMID: 38141160 DOI: 10.1080/17425255.2023.2298827] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/20/2023] [Indexed: 12/24/2023]
Abstract
INTRODUCTION The application of Artificial Intelligence (AI) to predictive toxicology is rapidly increasing, particularly aiming to develop non-testing methods that effectively address ethical concerns and reduce economic costs. In this context, Developmental Toxicity (Dev Tox) stands as a key human health endpoint, especially significant for safeguarding maternal and child well-being. AREAS COVERED This review outlines the existing methods employed in Dev Tox predictions and underscores the benefits of utilizing New Approach Methodologies (NAMs), specifically focusing on eXplainable Artificial Intelligence (XAI), which proves highly efficient in constructing reliable and transparent models aligned with recommendations from international regulatory bodies. EXPERT OPINION The limited availability of high-quality data and the absence of dependable Dev Tox methodologies render XAI an appealing avenue for systematically developing interpretable and transparent models, which hold immense potential for both scientific evaluations and regulatory decision-making.
Collapse
Affiliation(s)
- Maria Vittoria Togo
- Department of Pharmacy - Pharmaceutical Sciences, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Fabrizio Mastrolorito
- Department of Pharmacy - Pharmaceutical Sciences, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Angelica Orfino
- Department of Pharmacy - Pharmaceutical Sciences, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Elisabetta Anna Graps
- ARESS Puglia - Agenzia Regionale strategica per laSalute ed il Sociale, Presidenza della Regione Puglia", Bari, Italy
| | - Anna Rita Tondo
- Department of Pharmacy - Pharmaceutical Sciences, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Cosimo Damiano Altomare
- Department of Pharmacy - Pharmaceutical Sciences, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Fulvio Ciriaco
- Department of Chemistry, Universitá degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Daniela Trisciuzzi
- Department of Pharmacy - Pharmaceutical Sciences, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Orazio Nicolotti
- Department of Pharmacy - Pharmaceutical Sciences, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Nicola Amoroso
- Department of Pharmacy - Pharmaceutical Sciences, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
44
|
Zhang Z, Zhao L, Wang J, Wang C. A Hierarchical Graph Neural Network Framework for Predicting Protein-Protein Interaction Modulators With Functional Group Information and Hypergraph Structure. IEEE J Biomed Health Inform 2024; 28:4295-4305. [PMID: 38564358 DOI: 10.1109/jbhi.2024.3384238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Accurate prediction of small molecule modulators targeting protein-protein interactions (PPIMs) remains a significant challenge in drug discovery. Existing machine learning-based models rely on manual feature engineering, which is tedious and task-specific. Recently, deep learning models based on graph neural networks have made remarkable progress in molecular representation learning. However, many graph-based approaches ignore molecular hierarchical structure modeling guided by domain knowledge. In chemistry, the functional groups of a molecule determine its interaction with specific targets. Therefore, we propose a hierarchical graph neural network framework (called HiGPPIM) for predicting PPIMs by integrating atom-level and functional group-level features of molecules. HiGPPIM constructs atom-level and functional group-level graphs based on chemical knowledge and learns graph representations using graph attention networks. Furthermore, a hypergraph attention network is designed in HiGPPIM to aggregate and transform two-level graph information. We evaluate the performance of HiGPPIM on eight PPI families and two prediction tasks, namely PPIM identification and potency prediction. Experimental results demonstrate that HiGPPIM achieves state-of-the-art performance on both tasks and that using functional group information to guide PPIM prediction is effective.
Collapse
|
45
|
Seixas Feio JA, de Oliveira ECL, de Sales CDS, da Costa KS, e Lima AHL. Investigating molecular descriptors in cell-penetrating peptides prediction with deep learning: Employing N, O, and hydrophobicity according to the Eisenberg scale. PLoS One 2024; 19:e0305253. [PMID: 38870192 PMCID: PMC11175476 DOI: 10.1371/journal.pone.0305253] [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] [Received: 12/11/2023] [Accepted: 05/27/2024] [Indexed: 06/15/2024] Open
Abstract
Cell-penetrating peptides comprise a group of molecules that can naturally cross the lipid bilayer membrane that protects cells, sharing physicochemical and structural properties, and having several pharmaceutical applications, particularly in drug delivery. Investigations of molecular descriptors have provided not only an improvement in the performance of classifiers but also less computational complexity and an enhanced understanding of membrane permeability. Furthermore, the employment of new technologies, such as the construction of deep learning models using overfitting treatment, promotes advantages in tackling this problem. In this study, the descriptors nitrogen, oxygen, and hydrophobicity on the Eisenberg scale were investigated, using the proposed ConvBoost-CPP composed of an improved convolutional neural network with overfitting treatment and an XGBoost model with adjusted hyperparameters. The results revealed favorable to the use of ConvBoost-CPP, having as input nitrogen, oxygen, and hydrophobicity together with ten other descriptors previously investigated in this research line, showing an increase in accuracy from 88% to 91.2% in cross-validation and 82.6% to 91.3% in independent test.
Collapse
Affiliation(s)
- Juliana Auzier Seixas Feio
- Laboratório de Inteligência Computacional e Pesquisa Operacional, Campus Belém, Instituto de Tecnologia, Universidade Federal do Pará, Pará, Brazil
| | - Ewerton Cristhian Lima de Oliveira
- Laboratório de Inteligência Computacional e Pesquisa Operacional, Campus Belém, Instituto de Tecnologia, Universidade Federal do Pará, Pará, Brazil
- Instituto Tecnológico Vale, Belém, Pará, Brazil
| | - Claudomiro de Souza de Sales
- Laboratório de Inteligência Computacional e Pesquisa Operacional, Campus Belém, Instituto de Tecnologia, Universidade Federal do Pará, Pará, Brazil
| | - Kauê Santana da Costa
- Laboratório de Simulação Computacional, Campus Marechal Rondom, Instituto de Biodiversidade, Universidade Federal do Oeste do Pará, Santarém, Pará, Brazil
| | - Anderson Henrique Lima e Lima
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará, Belém, Pará, Brazil
| |
Collapse
|
46
|
Chen X, Huang J, Shen T, Zhang H, Xu L, Yang M, Xie X, Yan Y, Yan J. DEAttentionDTA: protein-ligand binding affinity prediction based on dynamic embedding and self-attention. Bioinformatics 2024; 40:btae319. [PMID: 38897656 PMCID: PMC11193059 DOI: 10.1093/bioinformatics/btae319] [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] [Received: 12/15/2023] [Revised: 03/23/2024] [Accepted: 06/17/2024] [Indexed: 06/21/2024] Open
Abstract
MOTIVATION Predicting protein-ligand binding affinity is crucial in new drug discovery and development. However, most existing models rely on acquiring 3D structures of elusive proteins. Combining amino acid sequences with ligand sequences and better highlighting active sites are also significant challenges. RESULTS We propose an innovative neural network model called DEAttentionDTA, based on dynamic word embeddings and a self-attention mechanism, for predicting protein-ligand binding affinity. DEAttentionDTA takes the 1D sequence information of proteins as input, including the global sequence features of amino acids, local features of the active pocket site, and linear representation information of the ligand molecule in the SMILE format. These three linear sequences are fed into a dynamic word-embedding layer based on a 1D convolutional neural network for embedding encoding and are correlated through a self-attention mechanism. The output affinity prediction values are generated using a linear layer. We compared DEAttentionDTA with various mainstream tools and achieved significantly superior results on the same dataset. We then assessed the performance of this model in the p38 protein family. AVAILABILITY AND IMPLEMENTATION The resource codes are available at https://github.com/whatamazing1/DEAttentionDTA.
Collapse
Affiliation(s)
- Xiying Chen
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jinsha Huang
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tianqiao Shen
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Houjin Zhang
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Li Xu
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Min Yang
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaoman Xie
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yunjun Yan
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jinyong Yan
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
47
|
Barone F, Russo ET, Villegas Garcia EN, Punta M, Cozzini S, Ansuini A, Cazzaniga A. Protein family annotation for the Unified Human Gastrointestinal Proteome by DPCfam clustering. Sci Data 2024; 11:568. [PMID: 38824125 PMCID: PMC11144186 DOI: 10.1038/s41597-024-03131-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 03/08/2024] [Indexed: 06/03/2024] Open
Abstract
Technological advances in massively parallel sequencing have led to an exponential growth in the number of known protein sequences. Much of this growth originates from metagenomic projects producing new sequences from environmental and clinical samples. The Unified Human Gastrointestinal Proteome (UHGP) catalogue is one of the most relevant metagenomic datasets with applications ranging from medicine to biology. However, the low levels of sequence annotation may impair its usability. This work aims to produce a family classification of UHGP sequences to facilitate downstream structural and functional annotation. This is achieved through the release of the DPCfam-UHGP50 dataset containing 10,778 putative protein families generated using DPCfam clustering, an unsupervised pipeline grouping sequences into single or multi-domain architectures. DPCfam-UHGP50 considerably improves family coverage at protein and residue levels compared to the manually curated repository Pfam. In the hope that DPCfam-UHGP50 will foster future discoveries in the field of metagenomics of the human gut, we release a FAIR-compliant database of our results that is easily accessible via a searchable web server and Zenodo repository.
Collapse
Affiliation(s)
- Federico Barone
- Area Science Park, Padriciano, 99, 34149, Trieste, Italy
- University of Trieste, Trieste, 34127, Italy
| | | | | | - Marco Punta
- IRCCS San Raffaele Institute, Center for Omics Sciences, Milan, 20132, Italy
- IRCCS San Raffaele Institute, Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, Milan, 20132, Italy
| | | | | | | |
Collapse
|
48
|
Jardim C, de Waal A, Fabris-Rotelli I, Rad NN, Mazarura J, Sherry D. Feature engineered embeddings for classification of molecular data. Comput Biol Chem 2024; 110:108056. [PMID: 38796282 DOI: 10.1016/j.compbiolchem.2024.108056] [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: 11/17/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 05/28/2024]
Abstract
The classification of molecules is of particular importance to the drug discovery process and several other use cases. Data in this domain can be partitioned into structural and sequence/text data. Several techniques such as deep learning are able to classify molecules and predict their functions using both types of data. Molecular structure and encoded chemical information are sufficient to classify a characteristic of a molecule. However, the use of a molecule's structural information typically requires large amounts of computational power with deep learning models that take a long time to train. In this study, we present an alternative approach to molecule classification that addresses the limitations of other techniques. This approach uses natural language processing techniques in the form of count vectorisation, term frequency-inverse document frequency, word2vec and Latent Dirichlet Allocation to feature engineer molecular text data. Through this approach, we aim to make a robust and easily reproducible embedding that is fast to implement and solely dependent on chemical (text) data such as the sequence of a protein. Further, we investigate the usefulness of these embeddings for machine learning models. We apply the techniques to two different types of molecular text data: FASTA sequence data and Simplified Molecular Input Line Entry Specification data. We show that these embeddings provide excellent performance for classification.
Collapse
|
49
|
Kadsanit N, Worsawat P, Sakonsinsiri C, McElroy CR, Macquarrie D, Noppawan P, Hunt AJ. Sustainable methods for the carboxymethylation and methylation of ursolic acid with dimethyl carbonate under mild and acidic conditions. RSC Adv 2024; 14:16921-16934. [PMID: 38799212 PMCID: PMC11124730 DOI: 10.1039/d4ra02122c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
Ursolic acid is a triterpene plant extract that exhibits significant potential as an anti-cancer, anti-tumour, and anti-inflammatory agent. Its direct use in the pharmaceutical industry is hampered by poor uptake of ursolic acid in the human body coupled with rapid metabolism causing a decrease in bioactivity. Modification of ursolic acid can overcome such issues, however, use of toxic reagents, unsustainable synthetic routes and poor reaction metrics have limited its potential. Herein, we demonstrate the first reported carboxymethylation and/or methylation of ursolic acid with dimethyl carbonate (DMC) as a green solvent and sustainable reagent under acidic conditions. The reaction of DMC with ursolic acid, in the presence of PTSA, ZnCl2, or H2SO4-SiO2 yielded the carboxymethylation product 3β-[[methoxy]carbonyl]oxyurs-12-en-28-oic acid, the methylation product 3β-methoxyurs-12-en-28-oic acid and the dehydration product urs-2,12-dien-28-oic acid. PTSA demonstrated high conversion and selectivity towards the previously unreported carboxymethylation of ursolic acid, while the application of formic acid in the system led to formylation of ursolic acid (3β-formylurs-12-en-28-oic acid) in quantitative yields via esterification, with DMC acting solely as a solvent. Meanwhile, the methylation product of ursolic acid, 3β-methoxyurs-12-en-28-oic acid, was successfully synthesised with FeCl3, demonstrating exceptional conversion and selectivity, >99% and 99%, respectively. Confirmed with the use of qualitative and quantitative green metrics, this result represents a significant improvement in conversion, selectivity, safety, and sustainability over previously reported methods of ursolic acid modification. It was demonstrated that these methods could be applied to other triterpenoids, including corosolic acid. The study also explored the potential pharmaceutical applications of ursolic acid, corosolic acid, and their derivatives, particularly in anti-inflammatory, anti-cancer, and anti-tumour treatments, using molecular ADMET and docking methods. The methods developed in this work have led to the synthesis of novel molecules, thus creating opportunities for the future investigation of biological activity and the modification of a wide range of triterpenoids applying acidic DMC systems to deliver novel active pharmaceutical intermediates.
Collapse
Affiliation(s)
- Nuttapong Kadsanit
- Materials Chemistry Research Center (MCRC), Department of Chemistry and Centre of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University Khon Kaen 40002 Thailand
| | - Pattamabhorn Worsawat
- Materials Chemistry Research Center (MCRC), Department of Chemistry and Centre of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University Khon Kaen 40002 Thailand
| | - Chadamas Sakonsinsiri
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University Khon Kaen 40002 Thailand
| | - Con R McElroy
- School of Chemistry, University of Lincoln Brayford Pool Campus Lincoln LN6 7TS UK
- Green Chemistry Centre of Excellence, Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Duncan Macquarrie
- Green Chemistry Centre of Excellence, Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Pakin Noppawan
- Department of Chemistry, Faculty of Science, Mahasarakham University Maha Sarakham 44150 Thailand
| | - Andrew J Hunt
- Materials Chemistry Research Center (MCRC), Department of Chemistry and Centre of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University Khon Kaen 40002 Thailand
| |
Collapse
|
50
|
Yoon JH, Lee D, Lee C, Cho E, Lee S, Cazenave-Gassiot A, Kim K, Chae S, Dennis EA, Suh PG. Paradigm shift required for translational research on the brain. Exp Mol Med 2024; 56:1043-1054. [PMID: 38689090 PMCID: PMC11148129 DOI: 10.1038/s12276-024-01218-x] [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: 10/13/2023] [Revised: 02/07/2024] [Accepted: 02/20/2024] [Indexed: 05/02/2024] Open
Abstract
Biomedical research on the brain has led to many discoveries and developments, such as understanding human consciousness and the mind and overcoming brain diseases. However, historical biomedical research on the brain has unique characteristics that differ from those of conventional biomedical research. For example, there are different scientific interpretations due to the high complexity of the brain and insufficient intercommunication between researchers of different disciplines owing to the limited conceptual and technical overlap of distinct backgrounds. Therefore, the development of biomedical research on the brain has been slower than that in other areas. Brain biomedical research has recently undergone a paradigm shift, and conducting patient-centered, large-scale brain biomedical research has become possible using emerging high-throughput analysis tools. Neuroimaging, multiomics, and artificial intelligence technology are the main drivers of this new approach, foreshadowing dramatic advances in translational research. In addition, emerging interdisciplinary cooperative studies provide insights into how unresolved questions in biomedicine can be addressed. This review presents the in-depth aspects of conventional biomedical research and discusses the future of biomedical research on the brain.
Collapse
Affiliation(s)
- Jong Hyuk Yoon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.
| | - Dongha Lee
- Cognitive Science Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Chany Lee
- Cognitive Science Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Eunji Cho
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Seulah Lee
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry and Precision Medicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
| | - Kipom Kim
- Research Strategy Office, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Sehyun Chae
- Neurovascular Unit Research Group, Korean Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Edward A Dennis
- Department of Pharmacology and Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0601, USA
| | - Pann-Ghill Suh
- Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| |
Collapse
|