1
|
Yoon D, Lee H. In silico discovery of novel compounds for FAK activation using virtual screening, AI-based prediction, and molecular dynamics. Comput Biol Chem 2025; 117:108420. [PMID: 40157227 DOI: 10.1016/j.compbiolchem.2025.108420] [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: 11/24/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 04/01/2025]
Abstract
Focal Adhesion Kinase (FAK) is a non-receptor tyrosine kinase that plays a crucial role in cell proliferation, migration, and signal transduction. FAK is overexpressed in metastatic and advanced-stage cancers, where it is considered a key kinase in cancer growth and metastasis. However, recent research has revealed that FAK activity decreases in various diseases. we aimed to identify compounds that could enhance FAK activity using structure-based virtual screening and artificial intelligence models from a vast chemical database. We began with an extensive chemical database containing over 10 million compounds and used our newly developed pipeline to screen candidate molecules. To select compounds structurally similar to ZINC40099027 (ZN27), a known FAK activator, we calculated Tanimoto Similarity scores and chose compounds with a score of at least 0.8. Clustering was performed using K-means based on the molecular properties. Subsequently, we utilized docking simulation, deep learning and SAScorer to evaluate and predict the protein-ligand docking affinity and physicochemical properties of the candidate compounds. The deep learning models were selected as state-of-the-art models: GLAM predicts the blood-brain barrier permeability of FAK, and elEmBERT predicts the potential toxicity of compound. The combined results were used to create an evaluation matrix. We selected 10 promising candidate compounds from the initial dataset of 10 million. To evaluate the stability of these top 10 candidate compounds in interaction with the FAK protein, we conducted Molecular Dynamics (MD) simulations. We performed a molecular dynamics simulation for a total of 50 ns and identified the top three promising candidate compounds.
Collapse
Affiliation(s)
- Deokhyeon Yoon
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hyunsu Lee
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
2
|
Ismail M, Liu J, Wang N, Zhang D, Qin C, Shi B, Zheng M. Advanced nanoparticle engineering for precision therapeutics of brain diseases. Biomaterials 2025; 318:123138. [PMID: 39914193 DOI: 10.1016/j.biomaterials.2025.123138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/31/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025]
Abstract
Despite the increasing global prevalence of neurological disorders, the development of nanoparticle (NP) technologies for brain-targeted therapies confronts considerable challenges. One of the key obstacles in treating brain diseases is the blood-brain barrier (BBB), which restricts the penetration of NP-based therapies into the brain. To address this issue, NPs can be installed with specific ligands or bioengineered to boost their precision and efficacy in targeting brain-diseased cells by navigating across the BBB, ultimately improving patient treatment outcomes. At the outset of this review, we highlighted the critical role of ligand-functionalized or bioengineered NPs in treating brain diseases from a clinical perspective. We then identified the key obstacles and challenges NPs encounter during brain delivery, including immune clearance, capture by the reticuloendothelial system (RES), the BBB, and the complex post-BBB microenvironment. Following this, we overviewed the recent progress in NPs engineering, focusing on ligand-functionalization or bionic designs to enable active BBB transcytosis and targeted delivery to brain-diseased cells. Lastly, we summarized the critical challenges hindering clinical translation, including scalability issues and off-target effects, while outlining future opportunities for designing cutting-edge brain delivery technologies.
Collapse
Affiliation(s)
- Muhammad Ismail
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Jiayi Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Ningyang Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Dongya Zhang
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Changjiang Qin
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China.
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW, 2109, Australia.
| | - Meng Zheng
- Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
3
|
Vidiyala N, Sunkishala P, Parupathi P, Nyavanandi D. The Role of Artificial Intelligence in Drug Discovery and Pharmaceutical Development: A Paradigm Shift in the History of Pharmaceutical Industries. AAPS PharmSciTech 2025; 26:133. [PMID: 40360908 DOI: 10.1208/s12249-025-03134-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
In today's world, with an increasing patient population, the need for medications is increasing rapidly. However, the current practice of drug development is time-consuming and requires a lot of investment by the pharmaceutical industries. Currently, it takes around 8-10 years and $3 billion of investment to develop a medication. Pharmaceutical industries and regulatory authorities are continuing to adopt new technologies to improve the efficiency of the drug development process. However, over the decades the pharmaceutical industries were not able to accelerate the drug development process. The pandemic (COVID-19) has taught the pharmaceutical industries and regulatory agencies an expensive lesson showing the need for emergency preparedness by accelerating the drug development process. Over the last few years, the pharmaceutical industries have been collaborating with artificial intelligence (AI) companies to develop algorithms and models that can be implemented at various stages of the drug development process to improve efficiency and reduce the developmental timelines significantly. In recent years, AI-screened drug candidates have entered clinical testing in human subjects which shows the interest of pharmaceutical companies and regulatory agencies. End-end integration of AI within the drug development process will benefit the industries for predicting the pharmacokinetic and pharmacodynamic profiles, toxicity, acceleration of clinical trials, study design, virtual monitoring of subjects, optimization of manufacturing process, analyzing and real-time monitoring of product quality, and regulatory preparedness. This review article discusses in detail the role of AI in various avenues of the pharmaceutical drug development process, its limitations, regulatory and future perspectives.
Collapse
Affiliation(s)
- Nithin Vidiyala
- Small Molecule Drug Product Development, Cerevel Therapeutics, Cambridge, Massachusetts, 02141, USA
| | - Pavani Sunkishala
- Process Validation, PCI Pharma Services, Bedford, New Hampshire, 03110, USA
| | - Prashanth Parupathi
- Division of Pharmaceutical Sciences, Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, New York, 11201, USA
| | - Dinesh Nyavanandi
- Small Molecule Drug Product Development, Cerevel Therapeutics, Cambridge, Massachusetts, 02141, USA.
| |
Collapse
|
4
|
Rybka H, Danel T, Podlewska S. PROFIS: Design of Target-Focused Libraries by Probing Continuous Fingerprint Space with Recurrent Neural Networks. J Chem Inf Model 2025; 65:4412-4425. [PMID: 40293047 DOI: 10.1021/acs.jcim.5c00698] [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: 04/30/2025]
Abstract
This study introduces PROFIS, a new generative model capable of the design of structurally novel and target-focused compound libraries. The model relies on a recurrent neural network that was trained to decode embedded molecular fingerprints into SMILES strings. To identify potential novel ligands, a biological activity predictor is first trained on the low-dimensional fingerprint embedding space, enabling the identification of high-activity subspaces for a given drug target. The search for latent representations that are expected to yield active structures upon decoding to SMILES is conducted with a Bayesian optimization algorithm. We present the rationale for using SMILES as the output notation of the recurrent neural network and compare its performance with models trained to decode DeepSMILES and SELFIES strings. The paper demonstrates the application of this protocol to generate candidate ligands of the dopamine D2 receptor. It also emphasizes the effectiveness of our approach in scaffold-hopping, which is valuable for designing ligands outside the already explored chemical space. We present how passing engineered molecular fingerprints through PROFIS network can be utilized to generate diverse libraries of analogs for a drug molecule of choice. It is worth noting that the protocol is versatile and it can be employed for any biological target, given the availability of a dataset containing known ligands. The potential for widespread use of PROFIS is secured by scripts shared by the authors on GitHub.
Collapse
Affiliation(s)
- Hubert Rybka
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Łojasiewicza 11, 30-348 Kraków, Poland
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Tomasz Danel
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Sabina Podlewska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smȩtna 12, 31-343 Kraków, Poland
| |
Collapse
|
5
|
Beilmann M, Adkins K, Boonen HCM, Hewitt P, Hu W, Mader R, Moore S, Rana P, Steger-Hartmann T, Villenave R, van Vleet T. Application of new approach methodologies for nonclinical safety assessment of drug candidates. Nat Rev Drug Discov 2025:10.1038/s41573-025-01182-9. [PMID: 40316753 DOI: 10.1038/s41573-025-01182-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2025] [Indexed: 05/04/2025]
Abstract
The development of new approach methodologies (NAMs) and advances with in vitro testing systems have prompted revisions in regulatory guidelines and inspired dedicated in vitro/ex vivo studies for nonclinical safety assessment. This Review by a safety reflection initiative subgroup of the European Federation of Pharmaceutical Industries and Associations (EFPIA)/Preclinical Development Expert Group (PDEG) summarizes the current state and potential application of in vitro studies using human-derived material for safety assessment in drug development. It focuses on case studies from recent projects in which animal models alone proved to be limited or inadequate for safety testing. It further highlights four categories of drug candidates for which alternative in vitro approaches are applicable and discusses progress in using in vitro testing solutions for safety assessment in these categories. Finally, the article highlights new risk assessment strategies, initiatives and consortia promoting the advancement of NAMs. This collective work is meant to encourage the use of NAMs for more human-relevant safety assessment, which should ultimately result in reduced animal testing for drug development.
Collapse
Affiliation(s)
- Mario Beilmann
- Global Nonclinical Safety & DMPK, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany.
| | | | | | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Wenyue Hu
- Vividion Therapeutics, San Diego, CA, USA
| | - Robert Mader
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Payal Rana
- Drug Safety R&D, Pfizer Inc., Groton, CT, USA
| | - Thomas Steger-Hartmann
- Research & Development, Pharmaceuticals, Preclinical Development, Bayer AG, Berlin, Germany
| | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | |
Collapse
|
6
|
Seal S, Mahale M, García-Ortegón M, Joshi CK, Hosseini-Gerami L, Beatson A, Greenig M, Shekhar M, Patra A, Weis C, Mehrjou A, Badré A, Paisley B, Lowe R, Singh S, Shah F, Johannesson B, Williams D, Rouquie D, Clevert DA, Schwab P, Richmond N, Nicolaou CA, Gonzalez RJ, Naven R, Schramm C, Vidler LR, Mansouri K, Walters WP, Wilk DD, Spjuth O, Carpenter AE, Bender A. Machine Learning for Toxicity Prediction Using Chemical Structures: Pillars for Success in the Real World. Chem Res Toxicol 2025. [PMID: 40314361 DOI: 10.1021/acs.chemrestox.5c00033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Machine learning (ML) is increasingly valuable for predicting molecular properties and toxicity in drug discovery. However, toxicity-related end points have always been challenging to evaluate experimentally with respect to in vivo translation due to the required resources for human and animal studies; this has impacted data availability in the field. ML can augment or even potentially replace traditional experimental processes depending on the project phase and specific goals of the prediction. For instance, models can be used to select promising compounds for on-target effects or to deselect those with undesirable characteristics (e.g., off-target or ineffective due to unfavorable pharmacokinetics). However, reliance on ML is not without risks, due to biases stemming from nonrepresentative training data, incompatible choice of algorithm to represent the underlying data, or poor model building and validation approaches. This might lead to inaccurate predictions, misinterpretation of the confidence in ML predictions, and ultimately suboptimal decision-making. Hence, understanding the predictive validity of ML models is of utmost importance to enable faster drug development timelines while improving the quality of decisions. This perspective emphasizes the need to enhance the understanding and application of machine learning models in drug discovery, focusing on well-defined data sets for toxicity prediction based on small molecule structures. We focus on five crucial pillars for success with ML-driven molecular property and toxicity prediction: (1) data set selection, (2) structural representations, (3) model algorithm, (4) model validation, and (5) translation of predictions to decision-making. Understanding these key pillars will foster collaboration and coordination between ML researchers and toxicologists, which will help to advance drug discovery and development.
Collapse
Affiliation(s)
- Srijit Seal
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Manas Mahale
- Department of Pharmaceutical Chemistry, Bombay College of Pharmacy, Mumbai 400098, India
| | | | - Chaitanya K Joshi
- Department of Computer Science and Technology, University of Cambridge, Cambridge CB3 0FD, U.K
| | | | - Alex Beatson
- Axiom Bio, San Francisco, California 94107, United States
| | - Matthew Greenig
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Mrinal Shekhar
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | | | | | | | - Adrien Badré
- Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Brianna Paisley
- Eli Lilly & Company, Indianapolis, Indiana 46285, United States
| | | | - Shantanu Singh
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Falgun Shah
- Non Clinical Drug Safety, Merck Inc., West Point, Pennsylvania 19486, United States
| | | | | | - David Rouquie
- Toxicology Data Science, Bayer SAS Crop Science Division, Valbonne Sophia-Antipolis 06560, France
| | - Djork-Arné Clevert
- Pfizer, Worldwide Research, Development and Medical, Machine Learning & Computational Sciences, Berlin 10922, Germany
| | | | | | - Christos A Nicolaou
- Computational Drug Design, Digital Science & Innovation, Novo Nordisk US R&D, Lexington, Massachusetts 02421, United States
| | - Raymond J Gonzalez
- Non Clinical Drug Safety, Merck Inc., West Point, Pennsylvania 19486, United States
| | - Russell Naven
- Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | | | | | - Kamel Mansouri
- NIH/NIEHS/DTT/NICEATM, Research Triangle Park, North Carolina 27709, United States
| | | | | | - Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala 751 24, Sweden
- Phenaros Pharmaceuticals AB, Uppsala 75239, Sweden
| | - Anne E Carpenter
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Andreas Bender
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| |
Collapse
|
7
|
Saif A, Islam MT, Raihan MO, Yousefi N, Rahman MA, Faridi H, Hasan AR, Hossain MM, Saleem RM, Albadrani GM, Al-Ghadi MQ, Ahasan Setu MA, Kamel M, Abdel-Daim MM, Aktaruzzaman M. Pan-cancer analysis of CDC7 in human tumors: Integrative multi-omics insights and discovery of novel marine-based inhibitors through machine learning and computational approaches. Comput Biol Med 2025; 190:110044. [PMID: 40120182 DOI: 10.1016/j.compbiomed.2025.110044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Cancer remains a significant global health challenge, with the Cell Division Cycle 7 (CDC7) protein emerging as a potential therapeutic target due to its critical role in tumor proliferation, survival, and resistance. However, a comprehensive analysis of CDC7 across multiple cancers is lacking, and existing therapeutic options have come with limited clinical success. The aim of this is to integrate a comprehensive pan-cancer analysis of CDC7 with the identification of novel marine-derived inhibitors, bridging the understanding of CDC7's role as a prognostic biomarker and therapeutic target across diverse cancer types. In this study, we conducted a pan-cancer analysis of CDC7 across 33 tumor types using publicly available datasets to evaluate its expression, genetic alterations, immune interactions, survival, and prognostic significance. Additionally, a marine-derived compound library of 31,492 molecules was screened to identify potential CDC7 inhibitors using chemoinformatics and machine learning. The top candidates underwent rigorous evaluations, including molecular docking, pharmacokinetics, toxicity, Density Functional Theory (DFT) calculations, and Molecular Dynamics (MD) simulations. The findings revealed that CDC7 is overexpressed in several cancers and is associated with poor survival outcomes and unfavorable prognosis. Enrichment analysis linked CDC7 to critical DNA replication pathways, while its role in modulating tumor-immune interactions highlighted its potential as a target for immunotherapy. Among all tested compounds, Tetrahydroaltersolanol D (CMNPD21999) exhibited the strongest binding affinity and stability, along with better drug-likeness and zero toxicity. These attributes highlight its potential as a promising drug candidate for CDC7 inhibition and future cancer treatment development. Furthermore, additional in vitro and in vivo studies are required to confirm the effectiveness of this drug candidate against the CDC7 protein.
Collapse
Affiliation(s)
- Ahmed Saif
- Department of Pharmacy, Faculty of Science, University of Rajshahi, Rajshahi, 6205, Bangladesh; Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh.
| | - Md Tarikul Islam
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh; Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh.
| | - Md Obayed Raihan
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Pharmaceutical Sciences, College of Health Sciences and Pharmacy, Chicago State University, Chicago, IL, USA.
| | - Niloofar Yousefi
- Department of Industrial Engineering and Management Systems, University of Central Florida, USA, Orlando, FL, USA
| | - Md Ajijur Rahman
- Department of Pharmacy, Faculty of Science, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Hafeez Faridi
- Department of Pharmaceutical Sciences, College of Health Sciences and Pharmacy, Chicago State University, Chicago, IL, USA
| | - Al Riyad Hasan
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Mirza Mahfuj Hossain
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Computer Science and Engineering, Faculty of Engineering and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Rasha Mohammed Saleem
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, 65431, Saudi Arabia
| | - Ghadeer M Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, 84428, Riyadh, 11671, Saudi Arabia
| | - Muath Q Al-Ghadi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Md Ali Ahasan Setu
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Microbiology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Mohamed M Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah, 21442, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Md Aktaruzzaman
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh.
| |
Collapse
|
8
|
Hamzyan Olia JB, Raman A, Hsu CY, Alkhayyat A, Nourazarian A. A comprehensive review of neurotransmitter modulation via artificial intelligence: A new frontier in personalized neurobiochemistry. Comput Biol Med 2025; 189:109984. [PMID: 40088712 DOI: 10.1016/j.compbiomed.2025.109984] [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: 11/05/2024] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
The deployment of artificial intelligence (AI) is revolutionizing neuropharmacology and drug development, allowing the modulation of neurotransmitter systems at the personal level. This review focuses on the neuropharmacology and regulation of neurotransmitters using predictive modeling, closed-loop neuromodulation, and precision drug design. The fusion of AI with applications such as machine learning, deep-learning, and even computational modeling allows for the real-time tracking and enhancement of biological processes within the body. An exemplary application of AI is the use of DeepMind's AlphaFold to design new GABA reuptake inhibitors for epilepsy and anxiety. Likewise, Benevolent AI and IBM Watson have fast-tracked drug repositioning for neurodegenerative conditions. Furthermore, we identified new serotonin reuptake inhibitors for depression through AI screening. In addition, the application of Deep Brain Stimulation (DBS) settings using AI for patients with Parkinson's disease and for patients with major depressive disorder (MDD) using reinforcement learning-based transcranial magnetic stimulation (TMS) leads to better treatment. This review highlights other challenges including algorithm bias, ethical concerns, and limited clinical validation. Their proposal to incorporate AI with optogenetics, CRISPR, neuroprosthesis, and other advanced technologies fosters further exploration and refinement of precision neurotherapeutic approaches. By bridging computational neuroscience with clinical applications, AI has the potential to revolutionize neuropharmacology and improve patient-specific treatment strategies. We addressed critical challenges, such as algorithmic bias and ethical concerns, by proposing bias auditing, diverse datasets, explainable AI, and regulatory frameworks as practical solutions to ensure equitable and transparent AI applications in neurotransmitter modulation.
Collapse
Affiliation(s)
| | - Arasu Raman
- Faculty of Business and Communications, INTI International University, Putra Nilai, 71800, Malaysia
| | - Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University, Tempe Campus, Phoenix, AZ, 85004, USA.
| | - Ahmad Alkhayyat
- Department of Computer Techniques Engineering, College of Technical Engineering, The Islamic University, Najaf, Iraq; Department of Computer Techniques Engineering, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran.
| |
Collapse
|
9
|
Alcaraz JML, Bouma H, Strodthoff N. Enhancing clinical decision support with physiological waveforms - A multimodal benchmark in emergency care. Comput Biol Med 2025; 192:110196. [PMID: 40311469 DOI: 10.1016/j.compbiomed.2025.110196] [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: 10/15/2024] [Revised: 03/04/2025] [Accepted: 04/09/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND AI-driven prediction algorithms have the potential to enhance emergency medicine by enabling rapid and accurate decision-making regarding patient status and potential deterioration. However, the integration of multimodal data, including raw waveform signals, remains underexplored in clinical decision support. METHODS We present a dataset and benchmarking protocol designed to advance multimodal decision support in emergency care. Our models utilize demographics, biometrics, vital signs, laboratory values, and electrocardiogram (ECG) waveforms as inputs to predict both discharge diagnoses and patient deterioration. RESULTS The diagnostic model achieves area under the receiver operating curve (AUROC) scores above 0.8 for 609 out of 1,428 conditions, covering both cardiac (e.g., myocardial infarction) and non-cardiac (e.g., renal disease, diabetes) diagnoses. The deterioration model attains AUROC scores above 0.8 for 14 out of 15 targets, accurately predicting critical events such as cardiac arrest, mechanical ventilation, ICU admission, and mortality. CONCLUSIONS Our study highlights the positive impact of incorporating raw waveform data into decision support models, improving predictive performance. By introducing a unique, publicly available dataset and baseline models, we provide a foundation for measurable progress in AI-driven decision support for emergency care.
Collapse
Affiliation(s)
- Juan Miguel Lopez Alcaraz
- AI4Health Division, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, Oldenburg, 26129, Lower Saxony, Germany.
| | - Hjalmar Bouma
- Department of Internal Medicine, Department of Acute Care, and Department of Clinical Pharmacy & Pharmacology, University Medical Center Groningen, Hanzeplein 1, Groningen, 9713, Groningen, Netherlands.
| | - Nils Strodthoff
- AI4Health Division, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, Oldenburg, 26129, Lower Saxony, Germany.
| |
Collapse
|
10
|
Ryzhkov FV, Ryzhkova YE, Elinson MN. Machine learning: Python tools for studying biomolecules and drug design. Mol Divers 2025:10.1007/s11030-025-11199-2. [PMID: 40301135 DOI: 10.1007/s11030-025-11199-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/13/2025] [Indexed: 05/01/2025]
Abstract
The increasing adoption of computational methods and artificial intelligence in scientific research has led to a growing interest in versatile tools like Python. In the fields of medical chemistry, biochemistry, and bioinformatics, Python has emerged as a key language for tackling complex challenges. It is used to solve various tasks, such as drug discovery, high-throughput and virtual screening, protein and genome analysis, and predicting drug efficacy. This review presents a list of tools for these tasks, including scripts, libraries, and ready-made programs, and serves as a starting point for scientists wishing to apply automation or optimization to routine tasks in medical chemistry and bioinformatics.
Collapse
Affiliation(s)
- Fedor V Ryzhkov
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991, Moscow, Russia.
| | - Yuliya E Ryzhkova
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991, Moscow, Russia
| | - Michail N Elinson
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991, Moscow, Russia
| |
Collapse
|
11
|
Li Z, Huang J, Liu X, Xu P, Shen X, Pan C, Zhang W, Liu W, Han H. KRN-DTI: Towards accurate drug-target interaction prediction with Kolmogorov-Arnold and residual networks. Methods 2025; 240:137-144. [PMID: 40287076 DOI: 10.1016/j.ymeth.2025.04.009] [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: 02/21/2025] [Revised: 04/03/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Predicting drug-target interactions (DTIs) accurately is essential in the field of drug discovery. Recently, artificial intelligence (AI) technologies, especially graph convolutional networks (GCNs), have been developed to tackle this challenge. However, as the number of GCN layers increases, models may lose critical information due to excessive smoothing. Moreover, these methods often lack interpretability and are dependent on specific datasets, which limits their generalizability. Consequently, this study introduces a novel method, KRN-DTI, which employs interpretable GCN technology to predict DTIs based on a drug-target heterogeneous network. The method uses GCN technology to identify potential DTIs by leveraging known interactions and dynamically adjusting the weights, thereby enhancing the model's interpretability. Additionally, residual connection technology is employed to integrate GNN outputs, mitigating the over-smoothing issue. Furthermore, the model's interpretability is enhanced by adaptively adjusting weights using Kolmogorov-Arnold Networks (KAN) and attention mechanisms. Experimental results show that KRN-DTI outperforms several advanced computational methods on the benchmark dataset. Case studies further highlight the effectiveness of KRN-DTI in predicting potential DTIs, showcasing its potential for real-world applications in drug discovery. Our code and data are publicly accessible at: https://github.com/lizhen5000/KRN-DTI.git.
Collapse
Affiliation(s)
- Zhen Li
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, Guangdong, 510006, China; School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, 558000, China
| | - Juyuan Huang
- Department of Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Xinxin Liu
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, Wenzhou, Zhejiang 325035, China.
| | - Peng Xu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, Guangdong, 510006, China.
| | - Xinwen Shen
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, Wenzhou, Zhejiang 325035, China
| | - Chu Pan
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan, 410000, China
| | - Wei Zhang
- Department of Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Wenbin Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, Guangdong, 510006, China.
| | - Henry Han
- School of engineering and computer science, Baylor University, Waco, TX, 76798, USA.
| |
Collapse
|
12
|
Mustoe CL, Turner AJ, Urwin SJ, Houson I, Feilden H, Markl D, Al Qaraghuli MM, Chong MWS, Robertson M, Nordon A, Johnston BF, Brown CJ, Robertson J, Adjiman C, Batchelor H, Benyahia B, Bresciani M, Burcham CL, Cardona J, Cottini C, Dunn AS, Fradet D, Halbert GW, Henson M, Hidber P, Langston M, Lee YS, Li W, Mantanus J, McGinty J, Mehta B, Naz T, Ottoboni S, Prasad E, Quist PO, Reynolds GK, Rielly C, Rowland M, Schlindwein W, Schroeder SLM, Sefcik J, Settanni E, Siddique H, Smith K, Smith R, Srai JS, Thorat AA, Vassileiou A, Florence AJ. Quality by digital design to accelerate sustainable medicines development. Int J Pharm 2025:125625. [PMID: 40287074 DOI: 10.1016/j.ijpharm.2025.125625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
We present a shared industry-academic perspective on the principles and opportunities for Quality by Digital Design (QbDD) as a framework to accelerate medicines development and enable regulatory innovation for new medicines approvals. This approach exploits emerging capabilities in industrial digital technologies to achieve robust control strategies assuring product quality and patient safety whilst reducing development time/costs, improving research and development efficiency, embedding sustainability into new products and processes, and promoting supply chain resilience. Key QbDD drivers include the opportunity for new scientific understanding and advanced simulation and model-driven, automated experimental approaches. QbDD accelerates the identification and exploration of more robust design spaces. Opportunities to optimise multiple objectives emerge in route selection, manufacturability and sustainability whilst assuring product quality. Challenges to QbDD adoption include siloed data and information sources across development stages, gaps in predictive capabilities, and the current extensive reliance on empirical knowledge and judgement. These challenges can be addressed via QbDD workflows; model-driven experimental design to collect and structure findable, accessible, interoperable and reusable (FAIR) data; and chemistry, manufacturing and control ontologies for shareable and reusable knowledge. Additionally, improved product, process, and performance predictive tools must be developed and exploited to provide a holistic end-to-end development approach.
Collapse
Affiliation(s)
- Chantal L Mustoe
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Alice J Turner
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Stephanie J Urwin
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Ian Houson
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Helen Feilden
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Daniel Markl
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Mohammed M Al Qaraghuli
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Magdalene W S Chong
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; WestCHEM, Department of Pure and Applied Chemistry and Centre for Process Analytics and Control Technology (CPACT), University of Strathclyde, Glasgow G1 1XL, United Kingdom
| | - Murray Robertson
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Alison Nordon
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; WestCHEM, Department of Pure and Applied Chemistry and Centre for Process Analytics and Control Technology (CPACT), University of Strathclyde, Glasgow G1 1XL, United Kingdom
| | - Blair F Johnston
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Cameron J Brown
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - John Robertson
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Claire Adjiman
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Department of Chemical Engineering, Sargent Centre for Process Systems Engineering, Imperial College London, London SW5 2AZ, United Kingdom
| | - Hannah Batchelor
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Brahim Benyahia
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Department of Chemical Engineering, Loughborough University, Loughborough LE11 3TU, United Kingdom
| | - Massimo Bresciani
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | | | - Javier Cardona
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Department of Chemical & Process Engineering, University of Strathclyde, Glasgow G1 1XJ, United Kingdom
| | | | - Andrew S Dunn
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - David Fradet
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Gavin W Halbert
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom; Cancer Research UK Formulation Unit, Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Mark Henson
- Takeda Pharmaceuticals International Co., Cambridge, MA 02139, USA
| | | | | | - Ye Seol Lee
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Department of Chemical Engineering, University College London, London WC1E 7JE, United Kingdom
| | - Wei Li
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Department of Chemical Engineering, Loughborough University, Loughborough LE11 3TU, United Kingdom
| | | | - John McGinty
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Department of Chemical & Process Engineering, University of Strathclyde, Glasgow G1 1XJ, United Kingdom
| | - Bhavik Mehta
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom; Siemens Industry Software Limited, London W6 7HA, United Kingdom
| | - Tabbasum Naz
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Sara Ottoboni
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Department of Chemical & Process Engineering, University of Strathclyde, Glasgow G1 1XJ, United Kingdom
| | - Elke Prasad
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Per-Ola Quist
- Operations, Pharmaceutical Technology & Development, Sustainable Innovation & Transformational Excellence (xSITE), AstraZeneca, Södertälje SE-151 85, Sweden
| | - Gavin K Reynolds
- Sustainable Innovation & Transformational Excellence (xSITE), Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield SK10 2NA, United Kingdom
| | - Chris Rielly
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Department of Chemical Engineering, Loughborough University, Loughborough LE11 3TU, United Kingdom
| | - Martin Rowland
- Pfizer Ltd., Discovery Park House, Sandwich, Kent CT13 9ND, United Kingdom
| | - Walkiria Schlindwein
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, United Kingdom
| | - Sven L M Schroeder
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, United Kingdom; Diamond Light Source, Didcot, Oxon OX11 0DE, United Kingdom
| | - Jan Sefcik
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Department of Chemical & Process Engineering, University of Strathclyde, Glasgow G1 1XJ, United Kingdom
| | - Ettore Settanni
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Institute for Manufacturing, Department of Engineering, University of Cambridge, Cambridge CB3 0FS, United Kingdom
| | - Humera Siddique
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Kenneth Smith
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Rachel Smith
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Jagjit Singh Srai
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Institute for Manufacturing, Department of Engineering, University of Cambridge, Cambridge CB3 0FS, United Kingdom
| | - Alpana A Thorat
- Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Antony Vassileiou
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Alastair J Florence
- CMAC, University of Strathclyde, Glasgow G1 1RD, United Kingdom; Strathclyde Institute of Pharmacy & Biomedical Science, University of Strathclyde, Glasgow G4 0RE, United Kingdom.
| |
Collapse
|
13
|
Smajić A, Steger-Hartmann T, Ecker GF, Hackl A. Data Exploration for Target Predictions Using Proprietary and Publicly Available Data Sets. Chem Res Toxicol 2025. [PMID: 40253625 DOI: 10.1021/acs.chemrestox.4c00347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
When applying machine learning (ML) approaches for the prediction of bioactivity, it is common to collect data from different assays or sources and combine them into single data sets. However, depending on the data domains and sources from which these data are retrieved, bioactivity data for the same macromolecular target may show a high variance of values (looking at a single compound) and cover very different parts of the chemical space as well as the bioactivity range (looking at the whole data set). The effectiveness and applicability domain of the resulting prediction models may be strongly influenced by the sources from which their training data were retrieved. Therefore, we investigated the chemical space and active/inactive distribution of proprietary pharmaceutical data from Bayer AG and the publicly available ChEMBL database, and their impact when applied as training data for classification models. For this end, we applied two different sets of descriptors in combination with different ML algorithms. The results show substantial differences in chemical space between the two different data sources, leading to suboptimal prediction performance when models are applied to domains other than their training data. MCC values between -0.34 and 0.37 among all targets were retrieved, indicating suboptimal model performance when models trained on Bayer AG data were tested on ChEMBL data and vice versa. The mean Tanimoto similarity of the nearest neighbors between these two data sources indicated similarities for 31 targets equal to or less than 0.3. Interestingly, all applied methods to assess overlap of chemical space of the two data sources to predict the applicability of models beyond their training data sets did not correlate with observed performances. Finally, we applied different strategies for creating mixed training data sets based on both public and proprietary sources, using assay format (cell-based and cell-free) information and Tanimoto similarities.
Collapse
Affiliation(s)
- Aljoša Smajić
- Department of Pharmaceutical Sciences, University of Vienna, Vienna 1090, Austria
| | | | - Gerhard F Ecker
- Department of Pharmaceutical Sciences, University of Vienna, Vienna 1090, Austria
| | - Anke Hackl
- Bayer AG, Pharmaceuticals Division, Berlin 13353, Germany
| |
Collapse
|
14
|
Ren J, Yan G, Yang L, Kong L, Guan Y, Sun H, Liu C, Liu L, Han Y, Wang X. Cancer chemoprevention: signaling pathways and strategic approaches. Signal Transduct Target Ther 2025; 10:113. [PMID: 40246868 PMCID: PMC12006474 DOI: 10.1038/s41392-025-02167-1] [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: 08/02/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025] Open
Abstract
Although cancer chemopreventive agents have been confirmed to effectively protect high-risk populations from cancer invasion or recurrence, only over ten drugs have been approved by the U.S. Food and Drug Administration. Therefore, screening potent cancer chemopreventive agents is crucial to reduce the constantly increasing incidence and mortality rate of cancer. Considering the lengthy prevention process, an ideal chemopreventive agent should be nontoxic, inexpensive, and oral. Natural compounds have become a natural treasure reservoir for cancer chemoprevention because of their superior ease of availability, cost-effectiveness, and safety. The benefits of natural compounds as chemopreventive agents in cancer prevention have been confirmed in various studies. In light of this, the present review is intended to fully delineate the entire scope of cancer chemoprevention, and primarily focuses on various aspects of cancer chemoprevention based on natural compounds, specifically focusing on the mechanism of action of natural compounds in cancer prevention, and discussing in detail how they exert cancer prevention effects by affecting classical signaling pathways, immune checkpoints, and gut microbiome. We also introduce novel cancer chemoprevention strategies and summarize the role of natural compounds in improving chemotherapy regimens. Furthermore, we describe strategies for discovering anticancer compounds with low abundance and high activity, revealing the broad prospects of natural compounds in drug discovery for cancer chemoprevention. Moreover, we associate cancer chemoprevention with precision medicine, and discuss the challenges encountered in cancer chemoprevention. Finally, we emphasize the transformative potential of natural compounds in advancing the field of cancer chemoprevention and their ability to introduce more effective and less toxic preventive options for oncology.
Collapse
Affiliation(s)
- Junling Ren
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Guangli Yan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Ling Kong
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Yu Guan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Hui Sun
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
| | - Chang Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Lei Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Ying Han
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Xijun Wang
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| |
Collapse
|
15
|
Cao K, Wang R, Wu S, Ou D, Li R, Li L, Liu X. Targeting Poly (ADP-ribose) polymerase-1 (PARP-1) for DNA repair mechanism through QSAR-based virtual screening and MD simulation. Mol Divers 2025:10.1007/s11030-025-11184-9. [PMID: 40227553 DOI: 10.1007/s11030-025-11184-9] [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: 02/18/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025]
Abstract
Poly (ADP-ribose) polymerase-1 (PARP-1) is a key enzyme in the base excision repair pathway, crucial for maintaining genomic stability by repairing DNA breaks. In cancers with mutations in DNA repair genes, such as BRCA1 and BRCA2, PARP-1 activity becomes essential for tumor cell survival, making it a promising target for therapeutic intervention. This study employs QSAR modeling, virtual screening, and molecular dynamics (MD) simulations to identify potential PARP-1 inhibitors. A dataset of inhibitors was analyzed using 12 molecular fingerprint descriptors to develop robust QSAR models, with the optimal model based on the CDK descriptor achieving R2 = 0.96, Q2_CV = 0.78, and Q2_Ext = 0.80. The model was applied to virtually screen three chemical libraries-ZINC, FDA, and NPA-identifying promising candidates for PARP-1 inhibition. Molecular docking revealed that compounds ZINC13132446, Z2037280227, and NPC193377 have strong binding affinity for the PARP-1 active site. MD simulations and MM-PBSA confirmed the stability of these complexes, with Z2037280227 and NPC193377 exhibiting the most stable interactions. These results underscore the potential of targeting PARP-1 as a therapeutic strategy for cancers with homologous recombination deficiencies, including prostate, breast, and ovarian cancer, particularly in patients with DNA repair deficiencies.
Collapse
Affiliation(s)
- Kun Cao
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China.
| | - Ruonan Wang
- Scientific Research Platform Management Service Center, Guangdong Medical University, Dongguan, 523808, China
| | - Siyu Wu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Dong Ou
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Ruixue Li
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Lianhai Li
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China.
| | - Xinguang Liu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
16
|
Aldakheel FM, Alduraywish SA, Dabwan KH. Integrating machine learning driven virtual screening and molecular dynamics simulations to identify potential inhibitors targeting PARP1 against prostate cancer. Sci Rep 2025; 15:12764. [PMID: 40229418 PMCID: PMC11997099 DOI: 10.1038/s41598-025-97208-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/02/2025] [Indexed: 04/16/2025] Open
Abstract
Prostate cancer (PC) is one of the most common types of malignancies in men, with a noteworthy increase in newly diagnosed cases in recent years. PARP1 is a ubiquitous nuclear enzyme involved in DNA repair, nuclear transport, ribosome synthesis, and epigenetic bookmarking. In this study, a library of 9000 phytochemicals was screened, with a focus on those with high drug efficacy and potential PARP1 inhibition. Different machine learning models were generated and assessed using various statistical measures. The RF model outperformed all other models in terms of accuracy (0.9489), specificity (0.9171), and area under the curve (AUC = 0.9846). Following this, a library of 9510 phytochemicals was screened, yielding 181 compounds predicted to be active. These compounds were subsequently assessed using Lipinski's Rule of Five, yielding 40 interesting candidates. Molecular docking experiments demonstrated that compound ZINC2356684563, ZINC2356558598, and ZINC14584870, had strong affinity for the PARP1 active site. Further molecular dynamics simulations and MM-PBSA validated the stability of the ligand-protein complexes, with ZINC14584870 and ZINC43120769 demonstrating the most stable interaction, as seen by low RMSD and RMSF levels. Our findings emphasize the potential of these phytochemical inhibitors as novel therapeutic agents against PARP1 in prostate cancer treatment, paving the path for further experimental validation and clinical investigations. These results open new possibilities for developing treatments to benefit prostate cancer patients.
Collapse
Affiliation(s)
- Fahad M Aldakheel
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, 11433, Riyadh, Saudi Arabia.
| | - Shatha A Alduraywish
- Department of Family and Community Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Khaled H Dabwan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, 11433, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Jia X, Teutonico D, Dhakal S, Psarellis YM, Abos A, Zhu H, Mavroudis PD, Pillai N. Application of Machine Learning and Mechanistic Modeling to Predict Intravenous Pharmacokinetic Profiles in Humans. J Med Chem 2025; 68:7737-7750. [PMID: 40146185 PMCID: PMC11998014 DOI: 10.1021/acs.jmedchem.5c00340] [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: 02/02/2025] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 03/28/2025]
Abstract
Accurate prediction of new compounds' pharmacokinetic (PK) profile in humans is crucial for drug discovery. Traditional methods, including allometric scaling and mechanistic modeling, rely on parameters from in vitro or in vivo testing, which are labor-intensive and involve ethical concerns. This study leverages machine learning (ML) to overcome these limitations by developing data-driven models. We compiled a large data set of small molecules' physicochemical and PK properties from public sources and digitized human plasma concentration-time profiles for approximately 800 compounds from the literature. We introduced a hybrid modeling framework that combines ML with physiologically based pharmacokinetic modeling and a hierarchical ML framework that employs two steps of learning to directly estimate PK profiles. Tested on 106 drugs, these frameworks demonstrated prediction accuracies within a 2-fold and 5-fold error for 40-60% and 80%-90% of compounds, respectively, in both AUC and Cmax. Proposed approaches could enhance early molecular screening and design, advancing drug discovery capabilities.
Collapse
Affiliation(s)
- Xuelian Jia
- Center
for Biomedical Informatics and Genomics, Tulane University, New Orleans, Louisiana 70112, United States
- Department
of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028, United States
| | - Donato Teutonico
- Quantitative
Pharmacology - Pharmacometrics, Sanofi, Vitry-sur-Seine 94400, France
| | - Saroj Dhakal
- Quantitative
Pharmacology - Pharmacometrics, Sanofi, Cambridge, Massachusetts 02141, United States
| | - Yorgos M. Psarellis
- Quantitative
Pharmacology - Pharmacometrics, Sanofi, Cambridge, Massachusetts 02141, United States
| | - Alexandra Abos
- Commercial
Data and Analytics, Sanofi, Barcelona 08016, Spain
| | - Hao Zhu
- Center
for Biomedical Informatics and Genomics, Tulane University, New Orleans, Louisiana 70112, United States
- Department
of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028, United States
| | - Panteleimon D. Mavroudis
- Quantitative
Pharmacology - Pharmacometrics, Sanofi, Cambridge, Massachusetts 02141, United States
| | - Nikhil Pillai
- Quantitative
Pharmacology - Pharmacometrics, Sanofi, Cambridge, Massachusetts 02141, United States
| |
Collapse
|
18
|
McGinnity DF, Meneyrol J, Boldron C, Johnstone C. Every Compound a Candidate: experience-led risk-taking approaches to accelerate small-molecule drug discovery. Drug Discov Today 2025; 30:104354. [PMID: 40209934 DOI: 10.1016/j.drudis.2025.104354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/12/2025]
Abstract
Despite progress, small-molecule drug discovery remains slow and costly. A paradigm shift is underway by leveraging artificial intelligence (AI) and machine learning (ML); however, these technological advances are necessary but not sufficient. Performance indicators from our partnered portfolio include timelines for data turnaround (5-day) and candidate delivery (2.9 versus 4.0 years for industry). Together with optimised processes and effective decision-making, improved translational predictivity is required. Progressing more compounds through downstream in vitro and in vivo models will rapidly reveal translational thresholds or crucial blockers for compound progression, with humans and machines actively learning from such data. We advocate for more experience-led risk-taking and a mindset shift toward an Every Compound a Candidate strategy, which aims to deliver drug candidates in <2 years.
Collapse
Affiliation(s)
- Dermot F McGinnity
- Aptuit (Verona) Srl, an Evotec Company, Via Alessandro Fleming 4, 37135 Verona, Italy.
| | | | | | | |
Collapse
|
19
|
Sutcliffe R, Doherty CPA, Morgan HP, Dunne NJ, McCarthy HO. Strategies for the design of biomimetic cell-penetrating peptides using AI-driven in silico tools for drug delivery. BIOMATERIALS ADVANCES 2025; 169:214153. [PMID: 39705787 DOI: 10.1016/j.bioadv.2024.214153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/08/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Cell-penetrating peptides (CPP) have gained rapid attention over the last 25 years; this is attributed to their versatility, customisation, and 'Trojan horse' delivery that evades the immune system. However, the current CPP rational design process is limited, as it requires several rounds of peptide synthesis, prediction and wet-lab validation, which is expensive, time-consuming and requires extensive knowledge in peptide chemistry. Artificial intelligence (AI) has emerged as a promising alternative which can augment the design process, for example by determining physiochemical characteristics, secondary structure, solvent accessibility, disorder and flexibility, as well as predicting in vivo behaviour such as toxicity and peptidase degradation. Other more recent tools utilise supervised machine learning (ML) to predict the penetrative ability of an amino acid sequence. The use of AI in the CPP design process has the potential to reduce development costs and increase the chances of success with respect to delivery. This review provides a survey of in silico tools and AI platforms which can be utilised in the design process, and the key features that should be taken into consideration when designing next generation CPPs.
Collapse
Affiliation(s)
- Rebecca Sutcliffe
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland
| | - Ciaran P A Doherty
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland; Antigenesis Biologics, Crossgar, Northern Ireland, United Kingdom of Great Britain and Northern Ireland
| | - Hugh P Morgan
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland; Antigenesis Biologics, Crossgar, Northern Ireland, United Kingdom of Great Britain and Northern Ireland
| | - Nicholas J Dunne
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland; School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland.
| |
Collapse
|
20
|
Shirzad M, Salahvarzi A, Razzaq S, Javid-Naderi MJ, Rahdar A, Fathi-Karkan S, Ghadami A, Kharaba Z, Romanholo Ferreira LF. Revolutionizing prostate cancer therapy: Artificial intelligence - Based nanocarriers for precision diagnosis and treatment. Crit Rev Oncol Hematol 2025; 208:104653. [PMID: 39923922 DOI: 10.1016/j.critrevonc.2025.104653] [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: 12/20/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/11/2025] Open
Abstract
Prostate cancer is one of the major health challenges in the world and needs novel therapeutic approaches to overcome the limitations of conventional treatment. This review delineates the transformative potential of artificial intelligence (AL) in enhancing nanocarrier-based drug delivery systems for prostate cancer therapy. With its ability to optimize nanocarrier design and predict drug delivery kinetics, AI has revolutionized personalized treatment planning in oncology. We discuss how AI can be integrated with nanotechnology to address challenges related to tumor heterogeneity, drug resistance, and systemic toxicity. Emphasis is placed on strong AI-driven advancements in the design of nanocarriers, structural optimization, targeting of ligands, and pharmacokinetics. We also give an overview of how AI can better predict toxicity, reduce costs, and enable personalized medicine. While challenges persist in the way of data accessibility, regulatory hurdles, and interactions with the immune system, future directions based on explainable AI (XAI) models, integration of multimodal data, and green nanocarrier designs promise to move the field forward. Convergence between AI and nanotechnology has been one key step toward safer, more effective, and patient-tailored cancer therapy.
Collapse
Affiliation(s)
- Maryam Shirzad
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsaneh Salahvarzi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sobia Razzaq
- School of Pharmacy, University of Management and Technology, Lahore SPH, Punjab, Pakistan
| | - Mohammad Javad Javid-Naderi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol, Iran.
| | - Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd 94531-55166, Iran; Department of Medical Nanotechnology, School of Medicine, North Khorasan University of Medical Science, Bojnurd, Iran.
| | - Azam Ghadami
- Department of Chemical and Polymer Engineering, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Zelal Kharaba
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | | |
Collapse
|
21
|
Dewaker V, Morya VK, Kim YH, Park ST, Kim HS, Koh YH. Revolutionizing oncology: the role of Artificial Intelligence (AI) as an antibody design, and optimization tools. Biomark Res 2025; 13:52. [PMID: 40155973 PMCID: PMC11954232 DOI: 10.1186/s40364-025-00764-4] [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: 02/25/2025] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
Antibodies play a crucial role in defending the human body against diseases, including life-threatening conditions like cancer. They mediate immune responses against foreign antigens and, in some cases, self-antigens. Over time, antibody-based technologies have evolved from monoclonal antibodies (mAbs) to chimeric antigen receptor T cells (CAR-T cells), significantly impacting biotechnology, diagnostics, and therapeutics. Although these advancements have enhanced therapeutic interventions, the integration of artificial intelligence (AI) is revolutionizing antibody design and optimization. This review explores recent AI advancements, including large language models (LLMs), diffusion models, and generative AI-based applications, which have transformed antibody discovery by accelerating de novo generation, enhancing immune response precision, and optimizing therapeutic efficacy. Through advanced data analysis, AI enables the prediction and design of antibody sequences, 3D structures, complementarity-determining regions (CDRs), paratopes, epitopes, and antigen-antibody interactions. These AI-powered innovations address longstanding challenges in antibody development, significantly improving speed, specificity, and accuracy in therapeutic design. By integrating computational advancements with biomedical applications, AI is driving next-generation cancer therapies, transforming precision medicine, and enhancing patient outcomes.
Collapse
Affiliation(s)
- Varun Dewaker
- Institute of New Frontier Research Team, Hallym University, Chuncheon-Si, Gangwon-Do, 24252, Republic of Korea
| | - Vivek Kumar Morya
- Department of Orthopedic Surgery, Hallym University Dongtan Sacred Hospital, Hwaseong-Si, 18450, Republic of Korea
| | - Yoo Hee Kim
- Department of Biomedical Gerontology, Ilsong Institute of Life Science, Hallym University, Seoul, 07247, Republic of Korea
| | - Sung Taek Park
- Institute of New Frontier Research Team, Hallym University, Chuncheon-Si, Gangwon-Do, 24252, Republic of Korea
- Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, 07441, Republic of Korea
- EIONCELL Inc, Chuncheon-Si, 24252, Republic of Korea
| | - Hyeong Su Kim
- Institute of New Frontier Research Team, Hallym University, Chuncheon-Si, Gangwon-Do, 24252, Republic of Korea.
- Department of Internal Medicine, Division of Hemato-Oncology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, 07441, Republic of Korea.
- EIONCELL Inc, Chuncheon-Si, 24252, Republic of Korea.
| | - Young Ho Koh
- Department of Biomedical Gerontology, Ilsong Institute of Life Science, Hallym University, Seoul, 07247, Republic of Korea.
| |
Collapse
|
22
|
Bobier C, Hurst DJ, Obeid J. Artificial intelligence, pharmaceutical development and dual-use research of concern: a call to action. JOURNAL OF MEDICAL ETHICS 2025:jme-2025-110750. [PMID: 40147882 DOI: 10.1136/jme-2025-110750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
Fervent attention was paid to what is coined dual-use research (DUR), or research that can both benefit and harm humanity, and dual-use research of concern (DURC), a particular subset of DUR that is reasonably anticipated to be a safety and security concern if misapplied. The aim of this paper is not to reiterate the challenges of DURC governance but to look at a new turn in DURC, namely the challenges posed by the use of artificial intelligence (AI) in pharmaceutical development. This is important, as AI is increasingly being used for pharmaceutical development in the industry. There is growing recognition that AI is DURC, and there is a dearth of industry and governmental guidance.
Collapse
Affiliation(s)
- Christopher Bobier
- Central Michigan University College of Medicine, Mount Pleasant, Michigan, USA
| | - Daniel J Hurst
- Director of Medical Professionalism, Ethics, & Humanities, Rowan-Virtua School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - John Obeid
- Central Michigan University College of Medicine, Mount Pleasant, Michigan, USA
| |
Collapse
|
23
|
Krüger FP, Östman J, Mervin L, Tetko IV, Engkvist O. Publishing neural networks in drug discovery might compromise training data privacy. J Cheminform 2025; 17:38. [PMID: 40140934 PMCID: PMC11948693 DOI: 10.1186/s13321-025-00982-w] [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: 12/10/2024] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
This study investigates the risks of exposing confidential chemical structures when machine learning models trained on these structures are made publicly available. We use membership inference attacks, a common method to assess privacy that is largely unexplored in the context of drug discovery, to examine neural networks for molecular property prediction in a black-box setting. Our results reveal significant privacy risks across all evaluated datasets and neural network architectures. Combining multiple attacks increases these risks. Molecules from minority classes, often the most valuable in drug discovery, are particularly vulnerable. We also found that representing molecules as graphs and using message-passing neural networks may mitigate these risks. We provide a framework to assess privacy risks of classification models and molecular representations, available at https://github.com/FabianKruger/molprivacy . Our findings highlight the need for careful consideration when sharing neural networks trained on proprietary chemical structures, informing organisations and researchers about the trade-offs between data confidentiality and model openness.
Collapse
Affiliation(s)
- Fabian P Krüger
- Discovery Sciences, Molecular AI, AstraZeneca R&D, Mölndal, 431 83, Sweden.
- TUM School of Computation, Information and Technology, Department of Mathematics, Technical University of Munich, Munich, 80333, Germany.
- Molecular Targets and Therapeutics Center, Institute of Structural Biology, Helmholtz Munich - Deutsches Forschungszentrum Für Gesundheit Und Umwelt (GmbH), Neuherberg, 85764, Germany.
| | | | - Lewis Mervin
- Discovery Sciences, Molecular AI, AstraZeneca R&D, Cambridge, CB2 0AA, UK
| | - Igor V Tetko
- Molecular Targets and Therapeutics Center, Institute of Structural Biology, Helmholtz Munich - Deutsches Forschungszentrum Für Gesundheit Und Umwelt (GmbH), Neuherberg, 85764, Germany
| | - Ola Engkvist
- Discovery Sciences, Molecular AI, AstraZeneca R&D, Mölndal, 431 83, Sweden
- Department of Computer Science and Engineering, Chalmers University of Technology, Gothenburg, 412 96, Sweden
| |
Collapse
|
24
|
Cassady H, Martin E, Liu Y, Bhattacharya D, Rochow MF, Dyer BA, Reinhart WF, Cooper VR, Hickner MA. Database of Nonaqueous Proton-Conducting Materials. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16901-16908. [PMID: 40059360 PMCID: PMC11931497 DOI: 10.1021/acsami.4c22618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/21/2025]
Abstract
This work presents the assembly of 48 papers, representing 74 different compounds and blends, into a machine-readable database of nonaqueous proton-conducting materials. SMILES was used to encode the chemical structures of the molecules, and we tabulated the reported proton conductivity, proton diffusion coefficient, and material composition for a total of 3152 data points. The data spans a broad range of temperatures ranging from -70 to 260 °C. To explore this landscape of nonaqueous proton conductors, DFT was used to calculate the proton affinity of 18 unique proton carriers. The results were then compared to the activation energy derived from fitting experimental data to the Arrhenius equation. It was found that while the widely recognized positive correlation between the activation energy and proton affinity may hold among closely related molecules, this correlation does not necessarily apply across a broader range of molecules. This work serves as an example of the potential analyses that can be conducted using literature data combined with emerging research tools in computation and data science to address specific materials design problems.
Collapse
Affiliation(s)
- Harrison
J. Cassady
- Department
of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan 48824-1312, United States
- Energy
Technologies Area, Lawrence Berkeley National
Laboratory, Berkeley 94720-8099, California, United States
| | - Emeline Martin
- Department
of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan 48824-1312, United States
- Department
of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109-1382, United
States
| | - Yifan Liu
- Materials
Science and Technology Division, Oak Ridge
National Laboratory, Oak Ridge, Tennessee 37831-2008, United States
| | - Debjyoti Bhattacharya
- Materials
Science and Engineering, The Pennsylvania
State University, University
Park, Pennsylvania 16802, United States
| | - Maria F. Rochow
- Department
of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan 48824-1312, United States
| | - Brock A. Dyer
- Department
of Physics and Astronomy, Ursinus College, Collegeville, Pennsylvania 19426, United States
| | - Wesley F. Reinhart
- Materials
Science and Engineering, The Pennsylvania
State University, University
Park, Pennsylvania 16802, United States
- Institute
for Computational and Data Sciences, The
Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Valentino R. Cooper
- Materials
Science and Technology Division, Oak Ridge
National Laboratory, Oak Ridge, Tennessee 37831-2008, United States
| | - Michael A. Hickner
- Department
of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan 48824-1312, United States
| |
Collapse
|
25
|
Cizauskas C, DeBenedictis E, Kelly P. How the past is shaping the future of life science: The influence of automation and AI on biology. N Biotechnol 2025; 88:1-11. [PMID: 40097138 DOI: 10.1016/j.nbt.2025.03.004] [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: 10/23/2024] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
Automation has been a transformative force for many industries, including manufacturing and chemistry. While the term traditionally referred to mechanical operations to produce physical objects, the definition has since expanded: 1) it can now mean both physical and/or information automation; and 2) it can now produce physical and/or conceptual outputs. While automation has yet to fully revolutionize life science research, much of which still relies on manual processes, we show that biology automation is the ultimate mixture of the concepts listed above - it involves automation of physical and data processing, and production of physical samples as well as conceptual data outputs. Here, we explore the history of automation and what it can - and cannot - teach us about the future of automated life science experimentation. We examine the current state of automated biology, its major successes, and the remaining barriers to wider adoption. Unlike in other fields, however, automation is reaching broader integration in life science at a time when both biology and AI are reaching their adolescence. At The Align Foundation, we are anticipating this change and hoping to leverage this inflection as a way to accelerate and democratize research. We anticipate that this novel combination of automation, AI, and life science learning will impact the trajectory of biological research, including the design and execution of high-throughput experiments and the analysis of resulting large-scale data.
Collapse
Affiliation(s)
| | | | - Pete Kelly
- The Align Foundation, Cambridge, MA 02138, United States.
| |
Collapse
|
26
|
Li J, Zhang J, Guo R, Dai J, Niu Z, Wang Y, Wang T, Jiang X, Hu W. Progress of machine learning in the application of small molecule druggability prediction. Eur J Med Chem 2025; 285:117269. [PMID: 39808972 DOI: 10.1016/j.ejmech.2025.117269] [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: 10/18/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Machine learning (ML) has become an important tool for predicting the pharmaceutical properties of small molecules. Recent advancements in ML algorithms enable the rapid and accurate evaluation of solubility, activity, toxicity, pharmacokinetics, and other molecular properties through ML-based models. By conducting virtual screening of drug targets and elucidating drug-target protein interactions, researchers can conduct preliminary evaluations of the activity and safety of compounds from the ultra-large drug compound libraries, thereby accelerating the screening process for lead compounds. Moreover, ML leverages existing experimental data to train and generate new datasets, addressing the challenge of limited compounds and protein target data. This review provided a concise overview of ML applications in predicting small molecule properties, focusing on model construction principles, molecular feature selection, and other essential aspects. It also discussed the potential applications of ML in the screening of pharmaceutical small molecules.
Collapse
Affiliation(s)
- Junyao Li
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China; School of Life Sciences, Huaiyin Normal University, Huaian, 223300, China; Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Jianmei Zhang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China
| | - Rui Guo
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China; Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Jiawei Dai
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Zhiqiang Niu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Yan Wang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China
| | - Taoyun Wang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China.
| | - Xiaojian Jiang
- School of Life Sciences, Huaiyin Normal University, Huaian, 223300, China.
| | - Weicheng Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
27
|
Guo W, Song X, Gao Y, Yang S, Tang J, Zhao C, Wang H, Ren J, Zeng L, Xu H. Exploring Insecticidal Molecules with Random Forest: Toward High Insecticidal Activity and Low Bee Toxicity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5573-5584. [PMID: 39978807 DOI: 10.1021/acs.jafc.4c08587] [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: 02/22/2025]
Abstract
Insecticidal molecules with high activity are crucial for global pesticide reduction and food security. However, their usage is limited by their concomitant high toxicity to bees. Balancing insecticidal activity and bee toxicity remains a critical challenge in the exploitation of new insecticidal molecules. In this study, we propose a novel strategy for exploiting molecules that are both highly effective against pests and minimally harmful to bees. A series of molecules were synthesized and tested to train a machine learning (ML) model for predicting insecticidal activity against pests. Meanwhile, another ML model was trained by using publicly available data to predict bee toxicity. The models demonstrated good performance, with mean AUC values of 0.88 ± 0.05 for insecticidal activity and 0.91 ± 0.01 for bee toxicity. By integrating these two models, we successfully predicted and experimentally validated a molecule that exhibited a high insecticidal activity and low bee toxicity. This dual-ML-model approach offers a promising pathway for the development of insecticidal molecules that are both effective and environmentally safe, thereby contributing to sustainable agricultures.
Collapse
Affiliation(s)
- Wei Guo
- State Key Laboratory of Green Pesticide, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, College of Plant Protection, South China Agricultural University, Guangzhou, Guangdong 510642, People's Republic of China
| | - Xiangmin Song
- State Key Laboratory of Green Pesticide, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, College of Plant Protection, South China Agricultural University, Guangzhou, Guangdong 510642, People's Republic of China
| | - Yongchao Gao
- State Key Laboratory of Green Pesticide, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, College of Plant Protection, South China Agricultural University, Guangzhou, Guangdong 510642, People's Republic of China
| | - Shuai Yang
- State Key Laboratory of Green Pesticide, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, College of Plant Protection, South China Agricultural University, Guangzhou, Guangdong 510642, People's Republic of China
| | - Jiahong Tang
- State Key Laboratory of Green Pesticide, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, College of Plant Protection, South China Agricultural University, Guangzhou, Guangdong 510642, People's Republic of China
| | - Chen Zhao
- State Key Laboratory of Green Pesticide, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, College of Plant Protection, South China Agricultural University, Guangzhou, Guangdong 510642, People's Republic of China
| | - Haojing Wang
- State Key Laboratory of Green Pesticide, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, College of Plant Protection, South China Agricultural University, Guangzhou, Guangdong 510642, People's Republic of China
| | - Jiajun Ren
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, 100875 Beijing, People's Republic of China
| | - Lingda Zeng
- State Key Laboratory of Green Pesticide, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, College of Plant Protection, South China Agricultural University, Guangzhou, Guangdong 510642, People's Republic of China
| | - Hanhong Xu
- State Key Laboratory of Green Pesticide, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, College of Plant Protection, South China Agricultural University, Guangzhou, Guangdong 510642, People's Republic of China
| |
Collapse
|
28
|
Rasul HO, Ghafour DD, Aziz BK, Hassan BA, Rashid TA, Kivrak A. Decoding Drug Discovery: Exploring A-to-Z In Silico Methods for Beginners. Appl Biochem Biotechnol 2025; 197:1453-1503. [PMID: 39630336 DOI: 10.1007/s12010-024-05110-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 03/29/2025]
Abstract
The drug development process is a critical challenge in the pharmaceutical industry due to its time-consuming nature and the need to discover new drug potentials to address various ailments. The initial step in drug development, drug target identification, often consumes considerable time. While valid, traditional methods such as in vivo and in vitro approaches are limited in their ability to analyze vast amounts of data efficiently, leading to wasteful outcomes. To expedite and streamline drug development, an increasing reliance on computer-aided drug design (CADD) approaches has merged. These sophisticated in silico methods offer a promising avenue for efficiently identifying viable drug candidates, thus providing pharmaceutical firms with significant opportunities to uncover new prospective drug targets. The main goal of this work is to review in silico methods used in the drug development process with a focus on identifying therapeutic targets linked to specific diseases at the genetic or protein level. This article thoroughly discusses A-to-Z in silico techniques, which are essential for identifying the targets of bioactive compounds and their potential therapeutic effects. This review intends to improve drug discovery processes by illuminating the state of these cutting-edge approaches, thereby maximizing the effectiveness and duration of clinical trials for novel drug target investigation.
Collapse
Affiliation(s)
- Hezha O Rasul
- Department of Pharmaceutical Chemistry, College of Science, Charmo University, Peshawa Street, Chamchamal, 46023, Sulaimani, Iraq.
| | - Dlzar D Ghafour
- Department of Medical Laboratory Science, College of Science, Komar University of Science and Technology, 46001, Sulaimani, Iraq
- Department of Chemistry, College of Science, University of Sulaimani, 46001, Sulaimani, Iraq
| | - Bakhtyar K Aziz
- Department of Nanoscience and Applied Chemistry, College of Science, Charmo University, Peshawa Street, Chamchamal, 46023, Sulaimani, Iraq
| | - Bryar A Hassan
- Computer Science and Engineering Department, School of Science and Engineering, University of Kurdistan Hewler, KRI, Iraq
- Department of Computer Science, College of Science, Charmo University, Peshawa Street, Chamchamal, 46023, Sulaimani, Iraq
| | - Tarik A Rashid
- Computer Science and Engineering Department, School of Science and Engineering, University of Kurdistan Hewler, KRI, Iraq
| | - Arif Kivrak
- Department of Chemistry, Faculty of Sciences and Arts, Eskisehir Osmangazi University, Eskişehir, 26040, Turkey
| |
Collapse
|
29
|
Brittin NJ, Anderson JM, Braun DR, Rajski SR, Currie CR, Bugni TS. Machine Learning-Based Bioactivity Classification of Natural Products Using LC-MS/MS Metabolomics. JOURNAL OF NATURAL PRODUCTS 2025; 88:361-372. [PMID: 39919314 DOI: 10.1021/acs.jnatprod.4c01123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
The rediscovery of known drug classes represents a major challenge in natural products drug discovery. Compound rediscovery inhibits the ability of researchers to explore novel natural products and wastes significant amounts of time and resources. This study introduces a novel machine learning framework that can effectively characterize the bioactivity of natural products by leveraging liquid chromatography tandem mass spectrometry and untargeted metabolomics analysis. This accelerates natural product drug discovery by addressing the challenge of dereplicating previously discovered bioactive compounds. Utilizing the SIRIUS 5 metabolomics software suite and in-silico-generated fragmentation spectra, we have trained a ML model capable of predicting a compound's drug class. This approach enables the rapid identification of bioactive scaffolds from LC-MS/MS data, even without reference experimental spectra. The model was trained on a diverse set of molecular fingerprints generated by SIRIUS 5 to effectively classify compounds based on their core pharmacophores. Our model robustly classified 21 diverse bioactive drug classes, achieving accuracies greater than 93% on experimental spectra. This study underscores the potential of ML combined with MFPs to dereplicate bioactive natural products based on pharmacophore, streamlining the discovery process and expediting improved methods of isolating novel antibacterial and antifungal agents.
Collapse
Affiliation(s)
- Nathaniel J Brittin
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Josephine M Anderson
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Doug R Braun
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Scott R Rajski
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Cameron R Currie
- Department of Biochemistry and Biomedical Sciences, M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Tim S Bugni
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Small Molecule Screening Facility, UW Carbone Cancer Center, Madison, Wisconsin 53792, United States
- Lachman Institute for Pharmaceutical Development, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
30
|
Kanwar RS, Gambhir K, Aggarwal T, Godiwal A, Bhadra K. From Spores to Suffering: Understanding the Role of Anthrax in Bioterrorism. Mil Med 2025; 190:e569-e579. [PMID: 39656926 DOI: 10.1093/milmed/usae535] [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: 06/25/2024] [Revised: 08/17/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
INTRODUCTION Anthrax, caused by the bacterium Bacillus anthracis, stands as a formidable threat with both natural and bioterrorism-related implications. Its ability to afflict a wide range of hosts, including humans and animals, coupled with its potential use as a bioweapon, underscores the critical importance of understanding and advancing our capabilities to combat this infectious disease. In this context, exploring futuristic approaches becomes imperative, as they hold the promise of not only addressing current challenges but also ushering in a new era in anthrax management. This review delves into strategies to mitigate the impact of anthrax on global health and security, envisioning a future where our arsenal against anthrax is characterized by precision and adaptability. MATERIALS AND METHODS This article highlights the significant potential of anthrax as a bioweapon, while also highlighting current knowledge and strategies developed against this deadly pathogen. We have performed an extensive research and literature review in concordance with the criteria outlined by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. A search strategy was conducted by using numerous keywords on various academic databases, yielding an initial set of 546 records along with 80 supplementary articles. The search included research papers, review papers, perspectives, clinical guidelines, and scientific blogs. The primary and secondary screening of the articles were conducted by 2 independent reviewers along with a third reviewer mitigating any discrepancies and biases encountered during the process. A set of inclusion and exclusion criteria were formulated, and a PICO framework was adapted based on which multiple records were analyzed and considered for the review. RESULTS In total, 53 articles were selected after completing a comprehensive systematic literature review. This review proposes novel approaches and scientific analysis of the complexities surrounding anthrax in the context of bioterrorism, highlighting the emerging technologies and strategies employed for bioterrorism mitigation. CONCLUSIONS The upcoming advancements in anthrax research will be based on cutting-edge technologies and innovative approaches that demonstrate great potential for prevention, detection, and treatment. These advancements may include the incorporation of synthetic biology techniques such as precise manipulation of biological components, nanoscale diagnostics, and Clustered regularly interspaced short palindromic repeats-based technologies, which could revolutionize our ability to combat anthrax on a molecular level. As these progressive methodologies continue to evolve, the integration of these technologies has the potential to redefine our strategies against anthrax, providing more accurate, personalized, and adaptable approaches to address the challenges posed by this infectious threat.
Collapse
Affiliation(s)
- Ratnesh Singh Kanwar
- Division of Clinical Research and Medical Management (CRMM), Institute of Nuclear Medicine & Allied Sciences (INMAS), DRDO, Delhi 110054, India
| | - Kirtida Gambhir
- Division of Clinical Research and Medical Management (CRMM), Institute of Nuclear Medicine & Allied Sciences (INMAS), DRDO, Delhi 110054, India
| | - Tanishka Aggarwal
- Division of Clinical Research and Medical Management (CRMM), Institute of Nuclear Medicine & Allied Sciences (INMAS), DRDO, Delhi 110054, India
| | - Akash Godiwal
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster 3rd Milestone, Faridabad, Haryana 121001, India
| | - Kuntal Bhadra
- Division of Clinical Research and Medical Management (CRMM), Institute of Nuclear Medicine & Allied Sciences (INMAS), DRDO, Delhi 110054, India
| |
Collapse
|
31
|
Cozac R, Hasic H, Choong JJ, Richard V, Beheshti L, Froehlich C, Koyama T, Matsumoto S, Kojima R, Iwata H, Hasegawa A, Otsuka T, Okuno Y. kMoL: an open-source machine and federated learning library for drug discovery. J Cheminform 2025; 17:22. [PMID: 40001146 PMCID: PMC11854109 DOI: 10.1186/s13321-025-00967-9] [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: 11/06/2024] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Machine learning is quickly becoming integral to drug discovery pipelines, particularly quantitative structure-activity relationship (QSAR) and absorption, distribution, metabolism, and excretion (ADME) tasks. Graph Convolutional Network (GCN) models have proven especially promising due to their inherent ability to model molecular structures using graph-based representations. However, maximizing the potential of such models in practice is challenging, as companies prioritize data privacy and security over collaboration initiatives to improve model performance and robustness. kMoL is an open-source machine learning library with integrated federated learning capabilities developed to address such challenges. Its key features include state-of-the-art model architectures, Bayesian optimization, explainability, and federated learning mechanisms. It demonstrates extensive customization possibilities, advanced security features, straightforward implementation of user-specific models, and high adaptability to custom datasets without additional programming requirements. kMoL is evaluated through locally trained benchmark settings and distributed federated learning experiments using various datasets to assess the features and flexibility of the library, as well as the ability to facilitate fast and practical experimentation. Additionally, results of these experiments provide further insights into the performance trade-offs associated with federated learning strategies, presenting valuable guidance for deploying machine learning models in a privacy-preserving manner within drug discovery pipelines. kMoL is available on GitHub at https://github.com/elix-tech/kmol .Scientific contribution The primary scientific contribution of this research project is the introduction and evaluation of kMoL, an open-source machine learning library with integrated federated learning capabilities. By demonstrating advanced customization and security capabilities without additional programming requirements, kMoL represents an accessible yet secure open-source platform for collaborative drug discovery projects. Additionally, the experiment results provide further insights into the performance trade-offs associated with federated learning strategies, presenting valuable guidance for deploying machine learning models in a privacy-preserving manner within drug discovery pipelines.
Collapse
Affiliation(s)
- Romeo Cozac
- Elix, Inc., 8-34 Yonbancho, Chiyoda-ku, Tokyo, 102-0081, Japan.
| | - Haris Hasic
- Elix, Inc., 8-34 Yonbancho, Chiyoda-ku, Tokyo, 102-0081, Japan
| | - Jun Jin Choong
- Elix, Inc., 8-34 Yonbancho, Chiyoda-ku, Tokyo, 102-0081, Japan
| | - Vincent Richard
- Elix, Inc., 8-34 Yonbancho, Chiyoda-ku, Tokyo, 102-0081, Japan
| | - Loic Beheshti
- Elix, Inc., 8-34 Yonbancho, Chiyoda-ku, Tokyo, 102-0081, Japan
| | | | - Takuto Koyama
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shigeyuki Matsumoto
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ryosuke Kojima
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroaki Iwata
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Aki Hasegawa
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takao Otsuka
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yasushi Okuno
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
32
|
Bachhar S, Kumar S, Dutta B, Das S. Emerging horizons of AI in pharmaceutical research. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2025; 103:325-348. [PMID: 40175048 DOI: 10.1016/bs.apha.2025.01.016] [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: 04/04/2025]
Abstract
Artificial Intelligence (AI) has revolutionized drug discovery by enhancing data collection, integration, and predictive modeling across various critical stages. It aggregates vast biological and chemical data, including genomic information, protein structures, and chemical interactions with biological targets. Machine learning techniques and QSAR models are applied by AI to predict compound behaviors and predict potential drug candidates. Docking simulations predict drug-protein interactions, while virtual screening eliminates large chemical databases through efficient sifting. Similarly, AI supports de novo drug design by generating novel molecules, optimized against a particular biological target, using generative models such as generative adversarial network (GAN), always finding lead compounds with the most desirable pharmacological properties. AI used in clinical trials improves efficiency by pinpointing responsive patient cohorts leveraging genetic profiles and biomarkers and maintaining propriety such as dataset diversity and compliance with regulations. This chapter aimed to summarize and analyze the mechanism of AI to accelerate drug discovery by streamlining different processes that enable informed decisions and bring potential life-saving therapies to market faster, amounting to a breakthrough in pharmaceutical research and development.
Collapse
Affiliation(s)
- Sourav Bachhar
- Department of Electronics and Communication Engineering, Kalyani Government Engineering College, Nadia, West Bengal, India; The Institute of Science Culture and Social Studies, Belgharia, Kolkata, West Bengal, India
| | - Suryasarathi Kumar
- The Institute of Science Culture and Social Studies, Belgharia, Kolkata, West Bengal, India; School of Biological Sciences & Technology, Department of Applied Biology, Maulana Abul Kalam Azad University of Technology, Haringhata, West Bengal, India
| | - Basudeb Dutta
- Institute for Integrated Cell-Material Sciences, Kyoto University, Yoshida Ushinomiya-cho, Sakyo-ku, Kyoto, Japan; Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India; Department of Chemistry, School of Applied Sciences, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha, India
| | - Somnath Das
- Department of Chemistry, School of Applied Sciences, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha, India.
| |
Collapse
|
33
|
Hayek-Orduz Y, Acevedo-Castro DA, Saldarriaga Escobar JS, Ortiz-Domínguez BE, Villegas-Torres MF, Caicedo PA, Barrera-Ocampo Á, Cortes N, Osorio EH, González Barrios AF. dyphAI dynamic pharmacophore modeling with AI: a tool for efficient screening of new acetylcholinesterase inhibitors. Front Chem 2025; 13:1479763. [PMID: 40017724 PMCID: PMC11865752 DOI: 10.3389/fchem.2025.1479763] [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: 08/12/2024] [Accepted: 01/06/2025] [Indexed: 03/01/2025] Open
Abstract
Therapeutic strategies for Alzheimer's disease (AD) often involve inhibiting acetylcholinesterase (AChE), underscoring the need for novel inhibitors with high selectivity and minimal side effects. A detailed analysis of the protein-ligand pharmacophore dynamics can facilitate this. In this study, we developed and employed dyphAI, an innovative approach integrating machine learning models, ligand-based pharmacophore models, and complex-based pharmacophore models into a pharmacophore model ensemble. This ensemble captures key protein-ligand interactions, including π-cation interactions with Trp-86 and several π-π interactions with residues Tyr-341, Tyr-337, Tyr-124, and Tyr-72. The protocol identified 18 novel molecules from the ZINC database with binding energy values ranging from -62 to -115 kJ/mol, suggesting their strong potential as AChE inhibitors. To further validate the predictions, nine molecules were acquired and tested for their inhibitory activity against human AChE. Experimental results revealed that molecules, 4 (P-1894047), with its complex multi-ring structure and numerous hydrogen bond acceptors, and 7 (P-2652815), characterized by a flexible, polar framework with ten hydrogen bond donors and acceptors, exhibited IC₅₀ values lower than or equal to that of the control (galantamine), indicating potent inhibitory activity. Similarly, molecules 5 (P-1205609), 6 (P-1206762), 8 (P-2026435), and 9 (P-533735) also demonstrated strong inhibition. In contrast, molecule 3 (P-617769798) showed a higher IC50 value, and molecules 1 (P-14421887) and 2 (P-25746649) yielded inconsistent results, likely due to solubility issues in the experimental setup. These findings underscore the value of integrating computational predictions with experimental validation, enhancing the reliability of virtual screening in the discovery of potent enzyme inhibitors.
Collapse
Affiliation(s)
- Yasser Hayek-Orduz
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá, Colombia
| | - Dorian Armando Acevedo-Castro
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá, Colombia
- Computational Bio-Organic Chemistry (COBO), Department of Chemistry, Universidad de los Andes, Bogotá, Colombia
| | - Juan Sebastián Saldarriaga Escobar
- Grupo Natura, Facultad de Ingenieria, Diseño y Ciencias Aplicadas, Departamento de Ciencias Biológicas, Bioprocesos y Biotecnología, Universidad ICESI, Cali, Colombia
| | - Brandon Eli Ortiz-Domínguez
- Grupo Natura, Facultad de Ingenieria, Diseño y Ciencias Aplicadas, Departamento de Ciencias Biológicas, Bioprocesos y Biotecnología, Universidad ICESI, Cali, Colombia
| | - María Francisca Villegas-Torres
- Centro de Investigaciones Microbiológicas (CIMIC), Department of Biological Sciences, Universidad de los Andes, Bogotá, Colombia
| | - Paola A. Caicedo
- Grupo Natura, Facultad de Ingenieria, Diseño y Ciencias Aplicadas, Departamento de Ciencias Biológicas, Bioprocesos y Biotecnología, Universidad ICESI, Cali, Colombia
| | - Álvaro Barrera-Ocampo
- Grupo Natura, Facultad de Ingenieria, Diseño y Ciencias Aplicadas, Departamento de Ciencias Farmacéuticas y Químicas, Universidad ICESI, Cali, Colombia
| | - Natalie Cortes
- Grupo de Investigación en Química Bioorgánica y Sistemas Moleculares (QBOSMO), Faculty of Natural Sciences and Mathematics, Universidad de Ibagué, Ibagué, Colombia
| | - Edison H. Osorio
- Grupo de Investigación en Química Bioorgánica y Sistemas Moleculares (QBOSMO), Faculty of Natural Sciences and Mathematics, Universidad de Ibagué, Ibagué, Colombia
| | - Andrés Fernando González Barrios
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá, Colombia
| |
Collapse
|
34
|
Albayati N, Talluri SR, Dholaria N, Michniak-Kohn B. AI-Driven Innovation in Skin Kinetics for Transdermal Drug Delivery: Overcoming Barriers and Enhancing Precision. Pharmaceutics 2025; 17:188. [PMID: 40006555 PMCID: PMC11859831 DOI: 10.3390/pharmaceutics17020188] [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: 12/21/2024] [Revised: 01/19/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Transdermal drug delivery systems (TDDS) offer an alternative to conventional oral and injectable drug administration by bypassing the gastrointestinal tract and liver metabolism, improving bioavailability, and minimizing systemic side effects. However, widespread adoption of TDDS is limited by challenges such as the skin's permeability barrier, particularly the stratum corneum, and the need for optimized formulations. Factors like skin type, hydration levels, and age further complicate the development of universally effective solutions. Advances in artificial intelligence (AI) address these challenges through predictive modeling and personalized medicine approaches. Machine learning models trained on extensive molecular datasets predict skin permeability and accelerate the selection of suitable drug candidates. AI-driven algorithms optimize formulations, including penetration enhancers and advanced delivery technologies like microneedles and liposomes, while ensuring safety and efficacy. Personalized TDDS design tailors drug delivery to individual patient profiles, enhancing therapeutic precision. Innovative systems, such as sensor-integrated patches, dynamically adjust drug release based on real-time feedback, ensuring optimal outcomes. AI also streamlines the pharmaceutical process, from disease diagnosis to the prediction of drug distribution in skin layers, enabling efficient formulation development. This review highlights AI's transformative role in TDDS, including applications of models such as Deep Neural Networks (DNN), Artificial Neural Networks (ANN), BioSIM, COMSOL, K-Nearest Neighbors (KNN), and Set Covering Machine (SVM). These technologies revolutionize TDDS for both skin and non-skin diseases, demonstrating AI's potential to overcome existing barriers and improve patient care through innovative drug delivery solutions.
Collapse
Affiliation(s)
- Nubul Albayati
- Ernest Mario School of Pharmacy, Rutgers-The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA; (N.A.); (S.R.T.); (N.D.)
- Center for Dermal Research, Rutgers-The State University of New Jersey, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Sesha Rajeswari Talluri
- Ernest Mario School of Pharmacy, Rutgers-The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA; (N.A.); (S.R.T.); (N.D.)
- Center for Dermal Research, Rutgers-The State University of New Jersey, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Nirali Dholaria
- Ernest Mario School of Pharmacy, Rutgers-The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA; (N.A.); (S.R.T.); (N.D.)
- Center for Dermal Research, Rutgers-The State University of New Jersey, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Bozena Michniak-Kohn
- Ernest Mario School of Pharmacy, Rutgers-The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA; (N.A.); (S.R.T.); (N.D.)
- Center for Dermal Research, Rutgers-The State University of New Jersey, 145 Bevier Road, Piscataway, NJ 08854, USA
| |
Collapse
|
35
|
Chung CK, Lin WY. ADR-DQPU: A Novel ADR Signal Detection Using Deep Reinforcement and Positive-Unlabeled Learning. IEEE J Biomed Health Inform 2025; 29:831-839. [PMID: 39499600 DOI: 10.1109/jbhi.2024.3492005] [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: 11/07/2024]
Abstract
The medical community has grappled with the challenge of analysis and early detection of severe and unknown adverse drug reactions (ADRs) from Spontaneous Reporting Systems (SRSs) like the FDA Adverse Event Reporting System (FAERS), which often lack professional verification and have inherent uncertainties. These limitations have exacerbated the difficulty of training a robust machine-learning model for detecting ADR signals from SRSs. A solution is to use some authoritative knowledge bases of ADRs, such as SIDER and BioSNAP, which contain limited confirmed ADR relationships (positive), resulting in a relatively small training set compared to the substantial amount of unknown data (unlabeled). This paper proposes a novel ADR signal detection method, ADR-DQPU, to alleviate the issues above by integrating deep reinforcement Q-learning and positive-unlabeled learning. Upon validation using FAERS data, our model outperformed six traditional methods, exhibiting an overall accuracy improvement of 26.45%, an average accuracy improvement of 52.15%, a precision enhancement of 1.89%, a recall improvement of 18.57%, and an F1 score improvement of 10.95%. In comparison to two state-of-the-art machine learning methods, our approach demonstrated an overall accuracy improvement of 64.1%, an average accuracy improvement of 28.23%, a slight decrease of 1.91% in precision, a recall improvement of 55.56%, and an F1 score improvement of 45.53%.
Collapse
|
36
|
Karimi-Sani I, Sharifi M, Abolpour N, Lotfi M, Atapour A, Takhshid MA, Sahebkar A. Drug repositioning for Parkinson's disease: An emphasis on artificial intelligence approaches. Ageing Res Rev 2025; 104:102651. [PMID: 39755176 DOI: 10.1016/j.arr.2024.102651] [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: 10/08/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
Parkinson's disease (PD) is one of the most incapacitating neurodegenerative diseases (NDDs). PD is the second most common NDD worldwide which affects approximately 1-2 percent of people over 65 years. It is an attractive pursuit for artificial intelligence (AI) to contribute to and evolve PD treatments through drug repositioning by repurposing existing drugs, shelved drugs, or even candidates that do not meet the criteria for clinical trials. A search was conducted in three databases Web of Science, Scopus, and PubMed. We reviewed the data related to the last years (1975-present) to identify those drugs currently being proposed for repositioning in PD. Moreover, we reviewed the present status of the computational approach, including AI/Machine Learning (AI/ML)-powered pharmaceutical discovery efforts and their implementation in PD treatment. It was found that the number of drug repositioning studies for PD has increased recently. Repositioning of drugs in PD is taking off, and scientific communities are increasingly interested in communicating its results and finding effective treatment alternatives for PD. A better chance of success in PD drug discovery has been made possible due to AI/ML algorithm advancements. In addition to the experimentation stage of drug discovery, it is also important to leverage AI in the planning stage of clinical trials to make them more effective. New AI-based models or solutions that increase the success rate of drug development are greatly needed.
Collapse
Affiliation(s)
- Iman Karimi-Sani
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehrdad Sharifi
- Emergency Medicine Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Nahid Abolpour
- Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehrzad Lotfi
- Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran; Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad-Ali Takhshid
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran; Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
37
|
Grover A, Singh S, Sindhu S, Lath A, Kumar S. Advances in cyclotide research: bioactivity to cyclotide-based therapeutics. Mol Divers 2025:10.1007/s11030-025-11113-w. [PMID: 39862350 DOI: 10.1007/s11030-025-11113-w] [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: 11/19/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025]
Abstract
Cyclotides are a class of plant-derived cyclic peptides having a distinctive structure with a cyclic cystine knot (CCK) motif. They are stable molecules that naturally play a role in plant defense. Till date, more than 750 cyclotides have been reported among diverse plant taxa belonging to Cucurbitaceae, Violaceae, Rubiaceae, Solanaceae, and Fabaceae. These native cyclotides exhibit several bioactivities, such as anti-bacterial, anti-HIV, anti-fungal, pesticidal, cytotoxic, and hemolytic activities which have immense significance in agriculture and therapeutics. The general mode of action of cyclotides is related to their structure, where their hydrophobic face penetrates the cell membrane and disrupts it to exhibit anti-microbial, cytotoxic, or hemolytic activities. Thus, the structure-activity relationship is of significance in cyclotides. Further, owing to their, small size, stability, and potential to interact and cross the membrane barrier of cells, they make promising choices for developing peptide-based biologics. However, challenges, such as production complexity, pharmacokinetic limitations, and off-target effects hinder their development. Advancements in cyclotide engineering, such as peptide grafting, ligand conjugation, and nanocarrier integration, heterologous production along with computational design optimization, can help overcome these challenges. Given the potential of these cyclic peptides, the present review focuses on the diversity, bioactivities, and structure-activity relationships of cyclotides, and advancements in cyclotides engineering emphasizing their unique attributes for diverse medical and biotechnological applications.
Collapse
Affiliation(s)
- Ankita Grover
- Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Sawraj Singh
- Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Sonal Sindhu
- Department of Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Amit Lath
- Department of Biotechnology, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Sanjay Kumar
- Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| |
Collapse
|
38
|
Hou Q, Li Y. Dual inhibition of AChE and MAO-B in Alzheimer's disease: machine learning approaches and model interpretations. Mol Divers 2025:10.1007/s11030-024-11061-x. [PMID: 39838228 DOI: 10.1007/s11030-024-11061-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/20/2024] [Indexed: 01/23/2025]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases. Given the multifactorial pathophysiology of AD, monotargeted agents can only alleviate symptoms but not cure AD. Acetylcholinesterase (AChE) and Monoamine oxidase B (MAO-B) are two key targets in the treatment of AD, molecules that inhibiting both targets are considered promising avenue to develop more effective AD therapies. In the present work, a dual inhibition dataset containing 449 molecules was established, based on which five machine learning algorithms (KNN, SVM, RF, GBDT, and LGBM) four fingerprints (MACCS, ECFP4, RDKitFP, PubChemFP) and DRAGON descriptors were combined to develop 25 classification models in which GBDT paired with ECFP4 and RF paired with PubchemFP achieved the same best performance across multiple metrics (Accuracy = 0.92, F1 Score = 0.94, MCC = 0.81). Moreover, based on the curated bioactivity datasets of AChE and MAO-B, regression models were developed to predict pIC50 values. For the AChE inhibition task, GBDT demonstrated the best performance (RMSE = 0.683, MAE = 0.500, R2 = 0.721). The SVM algorithm emerged as the most effective for MAO-B inhibition (RMSE = 0.668, MAE = 0.507, R2 = 0.675). The SHAP algorithm was used to interpret the optimal models, identifying and analyzing the key substructures and properties for both dual-target and single-target inhibitors. Moreover, molecules docking process provided potential mechanism and Structure-Activity Relationships (SAR) of dual-target inhibition further.
Collapse
Affiliation(s)
- Qinghe Hou
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, 116024, Liaoning, China
| | - Yan Li
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, 116024, Liaoning, China.
| |
Collapse
|
39
|
El-Tanani M, Rabbani SA, El-Tanani Y, Matalka II, Khalil IA. Bridging the gap: From petri dish to patient - Advancements in translational drug discovery. Heliyon 2025; 11:e41317. [PMID: 39811269 PMCID: PMC11730937 DOI: 10.1016/j.heliyon.2024.e41317] [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: 09/28/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Translational research serves as the bridge between basic research and practical applications in clinical settings. The journey from "bench to bedside" is fraught with challenges and complexities such as the often-observed disparity between how compounds behave in a laboratory setting versus in the complex systems of living organisms. The challenge is further compounded by the limited ability of in vitro models to mimic the specific biochemical environment of human tissues. This article explores and details the recent advancements and innovative approaches that are increasingly successful in bridging the gap between laboratory research and patient care. These advancements include, but are not limited to, sophisticated in vitro models such as organ-on-a-chip and computational models that utilize artificial intelligence to predict drug efficacy and safety. The article aims to showcase how these technologies improve the predictability of drug performance in human bodies and significantly speed up the drug development process. Furthermore, it discusses the role of biomarker discovery in preparation of more targeted and personalized therapy approaches and covers the impact of regulatory changes designed to facilitate drug approvals. Additionally, by providing detailed case studies of successful applications, we illustrate the practical impacts of these innovations on drug discovery and patient care.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Syed Arman Rabbani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | | | - Ismail I. Matalka
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Department of Pathology and Microbiology, Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Ikramy A. Khalil
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| |
Collapse
|
40
|
Cheng AH, Ser CT, Skreta M, Guzmán-Cordero A, Thiede L, Burger A, Aldossary A, Leong SX, Pablo-García S, Strieth-Kalthoff F, Aspuru-Guzik A. Spiers Memorial Lecture: How to do impactful research in artificial intelligence for chemistry and materials science. Faraday Discuss 2025; 256:10-60. [PMID: 39400305 DOI: 10.1039/d4fd00153b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Machine learning has been pervasively touching many fields of science. Chemistry and materials science are no exception. While machine learning has been making a great impact, it is still not reaching its full potential or maturity. In this perspective, we first outline current applications across a diversity of problems in chemistry. Then, we discuss how machine learning researchers view and approach problems in the field. Finally, we provide our considerations for maximizing impact when researching machine learning for chemistry.
Collapse
Affiliation(s)
- Austin H Cheng
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada.
- Department of Computer Science, University of Toronto, Toronto, Ontario M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
| | - Cher Tian Ser
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada.
- Department of Computer Science, University of Toronto, Toronto, Ontario M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
| | - Marta Skreta
- Department of Computer Science, University of Toronto, Toronto, Ontario M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
| | - Andrés Guzmán-Cordero
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
- Tinbergen Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Luca Thiede
- Department of Computer Science, University of Toronto, Toronto, Ontario M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
| | - Andreas Burger
- Department of Computer Science, University of Toronto, Toronto, Ontario M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
| | | | - Shi Xuan Leong
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada.
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 63737, Singapore
| | | | | | - Alán Aspuru-Guzik
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada.
- Department of Computer Science, University of Toronto, Toronto, Ontario M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario M5G 1M1, Canada
- Acceleration Consortium, Toronto, Ontario M5G 1X6, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada
- Department of Materials Science and Engineering, University of Toronto, Canada
- Lebovic Fellow, Canadian Institute for Advanced Research (CIFAR), Canada
| |
Collapse
|
41
|
Wellawatte GP, Schwaller P. Human interpretable structure-property relationships in chemistry using explainable machine learning and large language models. Commun Chem 2025; 8:11. [PMID: 39809811 PMCID: PMC11733140 DOI: 10.1038/s42004-024-01393-y] [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: 07/20/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
Explainable Artificial Intelligence (XAI) is an emerging field in AI that aims to address the opaque nature of machine learning models. Furthermore, it has been shown that XAI can be used to extract input-output relationships, making them a useful tool in chemistry to understand structure-property relationships. However, one of the main limitations of XAI methods is that they are developed for technically oriented users. We propose the XpertAI framework that integrates XAI methods with large language models (LLMs) accessing scientific literature to generate accessible natural language explanations of raw chemical data automatically. We conducted 5 case studies to evaluate the performance of XpertAI. Our results show that XpertAI combines the strengths of LLMs and XAI tools in generating specific, scientific, and interpretable explanations.
Collapse
Affiliation(s)
- Geemi P Wellawatte
- Laboratory of Artificial Chemical Intelligence, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Philippe Schwaller
- Laboratory of Artificial Chemical Intelligence, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- National Centre of Competence in Research (NCCR) Catalysis, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
42
|
DeCorte J, Brown B, Jeffrey R, Meiler J. Interpretable Deep-Learning p Ka Prediction for Small Molecule Drugs via Atomic Sensitivity Analysis. J Chem Inf Model 2025; 65:101-113. [PMID: 39801290 PMCID: PMC11733947 DOI: 10.1021/acs.jcim.4c01472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
Machine learning (ML) models now play a crucial role in predicting properties essential to drug development, such as a drug's logscale acid-dissociation constant (pKa). Despite recent architectural advances, these models often generalize poorly to novel compounds due to a scarcity of ground-truth data. Further, these models lack interpretability. To this end, with deliberate molecular embeddings, atomic-resolution information is accessible in chemical structures by observing the model response to atomic perturbations of an input molecule. Here, we present BCL-XpKa, a deep neural network (DNN)-based multitask classifier for pKa prediction that encodes local atomic environments through Mol2D descriptors. BCL-XpKa outputs a discrete distribution for each molecule, which stores the pKa prediction and the model's uncertainty for that molecule. BCL-XpKa generalizes well to novel small molecules. BCL-XpKa performs competitively with modern ML pKa predictors, outperforms several models in generalization tasks, and accurately models the effects of common molecular modifications on a molecule's ionizability. We then leverage BCL-XpKa's granular descriptor set and distribution-centered output through atomic sensitivity analysis (ASA), which decomposes a molecule's predicted pKa value into its respective atomic contributions without model retraining. ASA reveals that BCL-XpKa has implicitly learned high-resolution information about molecular substructures. We further demonstrate ASA's utility in structure preparation for protein-ligand docking by identifying ionization sites in 93.2% and 87.8% of complex small molecule acids and bases. We then applied ASA with BCL-XpKa to identify and optimize the physicochemical liabilities of a recently published KRAS-degrading PROTAC.
Collapse
Affiliation(s)
- Joseph DeCorte
- Department
of Chemical and Physical Biology, Vanderbilt
University, Nashville, Tennessee 37232, United States
- Center
for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt
Medical Scientist Training Program, Vanderbilt University Medical
Center, Vanderbilt University School of
Medicine, Nashville, Tennessee 37232-8725, United States
| | - Benjamin Brown
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232-8275, United
States
- Center
for Applied AI in Protein Dynamics, Vanderbilt
University, Nashville, Tennessee 37232-8725, United States
| | - Rathmell Jeffrey
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Jens Meiler
- Department
of Chemical and Physical Biology, Vanderbilt
University, Nashville, Tennessee 37232, United States
- Center
for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232-8275, United
States
- Center
for Applied AI in Protein Dynamics, Vanderbilt
University, Nashville, Tennessee 37232-8725, United States
- Institute
for Drug Discovery, Leipzig University Medical
School, Leipzig, SAC 04103, Germany
| |
Collapse
|
43
|
Zhang Y, Vitalis A. Benchmarking the robustness of the correct identification of flexible 3D objects using common machine learning models. PATTERNS (NEW YORK, N.Y.) 2025; 6:101147. [PMID: 39896260 PMCID: PMC11783895 DOI: 10.1016/j.patter.2024.101147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/09/2024] [Accepted: 12/10/2024] [Indexed: 02/04/2025]
Abstract
True three-dimensional (3D) data are prevalent in domains such as molecular science or computer vision. In these data, machine learning models are often asked to identify objects subject to intrinsic flexibility. Our study introduces two datasets from molecular science to assess the classification robustness of common model/feature combinations. Molecules are flexible, and shapes alone offer intra-class heterogeneities that yield a high risk for confusions. By blocking training and test sets to reduce overlap, we establish a baseline requiring the trained models to abstract from shape. As training data coverage grows, all tested architectures perform better on unseen data with reduced overfitting. Empirically, 2D embeddings of voxelized data produced the best-performing models. Evidently, both featurization and task-appropriate model design are of continued importance, the latter point reinforced by comparisons to recent, more specialized models. Finally, we show that the shape abstraction learned from database samples extends to samples that are evolving explicitly in time.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Andreas Vitalis
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
44
|
Rosa D, Elya B, Hanafi M, Khatib A, Budiarto E, Nur S, Surya MI. Investigation of alpha-glucosidase inhibition activity of Artabotrys sumatranus leaf extract using metabolomics, machine learning and molecular docking analysis. PLoS One 2025; 20:e0313592. [PMID: 39752479 PMCID: PMC11698457 DOI: 10.1371/journal.pone.0313592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/27/2024] [Indexed: 01/06/2025] Open
Abstract
One way to treat diabetes mellitus type II is by using α-glucosidase inhibitor, that will slow down the postprandial glucose intake. Metabolomics analysis of Artabotrys sumatranus leaf extract was used in this research to predict the active compounds as α-glucosidase inhibitors from this extract. Both multivariate statistical analysis and machine learning approaches were used to improve the confidence of the predictions. After performance comparisons with other machine learning methods, random forest was chosen to make predictive model for the activity of the extract samples. Feature importance analysis (using random feature permutation and Shapley score calculation) was used to identify the predicted active compound as the important features that influenced the activity prediction of the extract samples. The combined analysis of multivariate statistical analysis and machine learning predicted 9 active compounds, where 6 of them were identified as mangiferin, neomangiferin, norisocorydine, apigenin-7-O-galactopyranoside, lirioferine, and 15,16-dihydrotanshinone I. The activities of norisocorydine, apigenin-7-O-galactopyranoside, and lirioferine as α-glucosidase inhibitors have not yet reported before. Molecular docking simulation, both to 3A4A (α-glucosidase enzyme from Saccharomyces cerevisiae, usually used in bioassay test) and 3TOP (a part of α-glucosidase enzyme in human gut) showed strong to very strong binding of the identified predicted active compounds to both receptors, with exception of neomangiferin which only showed strong binding to 3TOP receptor. Isolation based on bioassay guided fractionation further verified the metabolomics prediction by succeeding to isolate mangiferin from the extract, which showed strong α-glucosidase activity when subjected to bioassay test. The correlation analysis also showed a possibility of 3 groups in the predicted active compounds, which might be related to the biosynthesis pathway (need further research for verification). Another result from correlation analysis was that in general the α-glucosidase inhibition activity in the extract had strong correlation to antioxidant activity, which was also reflected in the predicted active compounds. Only one predicted compound had very low positive correlation to antioxidant activity.
Collapse
Affiliation(s)
- Dela Rosa
- Department of Pharmacy, Faculty of Pharmacy, Indonesia University, Depok, Indonesia
- Department of Pharmacy, Faculty of Health Science, Pelita Harapan University, Tangerang, Indonesia
| | - Berna Elya
- Department of Pharmacy, Faculty of Pharmacy, Indonesia University, Depok, Indonesia
| | - Muhammad Hanafi
- Chemistry Research Centre, National Research and Innovation Agency, Science and Technology Research Centre, Serpong, Indonesia
| | - Alfi Khatib
- Department of Pharmaceutical Chemistry, Kulliyah of Pharmacy, International Islamic University Malaysia, Kuantan, Malaysia
| | - Eka Budiarto
- Department of Information Technology, Faculty of Engineering and Information Technology, Swiss German University, Tangerang, Indonesia
| | - Syamsu Nur
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Almarisah Madani University, Makasar, Indonesia
| | - Muhammad Imam Surya
- Research Centre for Plant Conservation, Botanic Gardens and Forestry, National Research and Innovation Agency, Bogor, Indonesia
| |
Collapse
|
45
|
Gupta M, Mukherjee T, Mohanty S. Accelerating Rheumatoid Arthritis Drug Repurposing: A Computational Approach. Curr Comput Aided Drug Des 2025; 21:125-128. [PMID: 39108125 DOI: 10.2174/0115734099326517240801035901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/29/2024] [Accepted: 07/08/2024] [Indexed: 04/05/2025]
Affiliation(s)
- Muskan Gupta
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Tuhin Mukherjee
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Satyajit Mohanty
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| |
Collapse
|
46
|
Bowyer S, Allen DJ, Furnham N. Unveiling the ghost: machine learning's impact on the landscape of virology. J Gen Virol 2025; 106. [PMID: 39804261 DOI: 10.1099/jgv.0.002067] [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: 05/02/2025] Open
Abstract
The complexity and speed of evolution in viruses with RNA genomes makes predictive identification of variants with epidemic or pandemic potential challenging. In recent years, machine learning has become an increasingly capable technology for addressing this challenge, as advances in methods and computational power have dramatically improved the performance of models and led to their widespread adoption across industries and disciplines. Nascent applications of machine learning technology to virus research have now expanded, providing new tools for handling large-scale datasets and leading to a reshaping of existing workflows for phenotype prediction, phylogenetic analysis, drug discovery and more. This review explores how machine learning has been applied to and has impacted the study of viruses, before addressing the strengths and limitations of its techniques and finally highlighting the next steps that are needed for the technology to reach its full potential in this challenging and ever-relevant research area.
Collapse
Affiliation(s)
- Sebastian Bowyer
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - David J Allen
- Department of Comparative Biomedical Sciences, Section Infection and Immunity, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Nicholas Furnham
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
47
|
Singh PK, Sachan K, Khandelwal V, Singh S, Singh S. Role of Artificial Intelligence in Drug Discovery to Revolutionize the Pharmaceutical Industry: Resources, Methods and Applications. Recent Pat Biotechnol 2025; 19:35-52. [PMID: 39840410 DOI: 10.2174/0118722083297406240313090140] [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: 12/07/2023] [Revised: 02/22/2024] [Accepted: 02/28/2024] [Indexed: 01/23/2025]
Abstract
Traditional drug discovery methods such as wet-lab testing, validations, and synthetic techniques are time-consuming and expensive. Artificial Intelligence (AI) approaches have progressed to the point where they can have a significant impact on the drug discovery process. Using massive volumes of open data, artificial intelligence methods are revolutionizing the pharmaceutical industry. In the last few decades, many AI-based models have been developed and implemented in many areas of the drug development process. These models have been used as a supplement to conventional research to uncover superior pharmaceuticals expeditiously. AI's involvement in the pharmaceutical industry was used mostly for reverse engineering of existing patents and the invention of new synthesis pathways. Drug research and development to repurposing and productivity benefits in the pharmaceutical business through clinical trials. AI is studied in this article for its numerous potential uses. We have discussed how AI can be put to use in the pharmaceutical sector, specifically for predicting a drug's toxicity, bioactivity, and physicochemical characteristics, among other things. In this review article, we have discussed its application to a variety of problems, including de novo drug discovery, target structure prediction, interaction prediction, and binding affinity prediction. AI for predicting drug interactions and nanomedicines were also considered.
Collapse
Affiliation(s)
- Pranjal Kumar Singh
- Department of Pharmacy, Kalka Institute for Research and Advanced Studies, Meerut, Uttar Pradesh, India
| | - Kapil Sachan
- KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India
| | - Vishal Khandelwal
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Sumita Singh
- Faculty of Pharmacy, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Smita Singh
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology, Delhi NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
48
|
Zhou S, Ye L, Huang Y, Valsecchi C, Liu Y, Shao L, Liu J, He T, Liu L, Fan M. Rapid diagnosis and recurrence prediction of choledocholithiasis disease using raw bile with machine learning assisted SERS. Talanta 2025; 282:126979. [PMID: 39383718 DOI: 10.1016/j.talanta.2024.126979] [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] [Revised: 09/06/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Surface-enhanced Raman spectroscopy (SERS) analysis based on body fluids has been widely applied in disease diagnose. Choledocholithiasis is a widespread and often recurrent digestive system disease, with limited data on factors predicting its formation and reappearance. Bile contains many components that could provide valuable diagnostic information; however, the current diagnosis of biliary disease by SERS focuses on detecting specific component in the bile, overlooking the complex interplay and correlations among multiple factors that could be crucial for accurate diagnosis. This work directly obtained multi-component SERS spectral information of raw bile from 46 patients. Characteristic information was extracted from bile SERS spectra using Principal Component Analysis (PCA), revealing variations in the content of characteristic components associated with different choledocholithiasis types and their recurrence frequency. Pearson correlation analysis was also introduced to reveal the interactions of primary active substances pertinent to choledocholithiasis diagnosis. The efficacy of PCA and Support Vector Machine (SVM) models in classifying stone types, presented an accuracy of 99.2 %. Furthermore, the interaction patterns among SERS characteristic components in choledocholithiasis recurrence frequency were revealed, and with the support of SVM, the prediction for different recurrence rates reached an accuracy of 95.2 %. Overall, this work demonstrates that integrating SERS with machine learning can support disease diagnosis and the interpretation of correlations among multiple components, facilitating elucidating the disease mechanisms.
Collapse
Affiliation(s)
- Shana Zhou
- School of Environmental Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Liansong Ye
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yuting Huang
- Department of Clinical Laboratory Medicine, First Affiliated Hospital, Army Medical University, Chongqing, 400038, China
| | - Chiara Valsecchi
- Federal University of Pampa, Campus Alegrete, 97542-160, Alegrete, RS, Brazil
| | - Yingying Liu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Limei Shao
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Jiao Liu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Tian He
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Ling Liu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China.
| | - Meikun Fan
- School of Environmental Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China.
| |
Collapse
|
49
|
Uppathi P, Rajakumari S, Saritha KV. Molecular Docking: An Emerging Tool for Target-Based Cancer Therapy. Crit Rev Oncog 2025; 30:1-13. [PMID: 39819431 DOI: 10.1615/critrevoncog.2024056533] [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: 01/19/2025]
Abstract
Molecular docking is a structure-based computational technique that plays a major role in drug discovery. Molecular docking enhances the efficacy of determining the metabolic interaction between two molecules, i.e., the small molecule (ligand) and the target molecule (protein), to find the best orientation of a ligand to its target molecule with minimal free energy in forming a stable complex. By stimulating drug-target interactions, docking helps identify small molecules that might inhibit cancer-promoting proteins, aiding in the development of novel targeted therapies. Molecular docking enables researchers to screen vast reorganization, identifying potential anti-cancer drugs with enhanced specificity and reduced toxicity. The growing importance of molecular docking underscores its potential to revolutionize cancer treatment by accelerating the identification of novel drugs and improving clinical outcomes. As a wide approach, this computational drug design technique can be considered more effective and timesaving than other cancer treatment methods. In this review, we showcase brief information on the role of molecular docking and its importance in cancer research for drug discovery and target identification. Therefore, in recent years, it can be concluded that molecular docking can be scrutinized as one of the novel strategies at the leading edge of cancer-targeting drug discovery.
Collapse
Affiliation(s)
| | - Suraj Rajakumari
- Department of Biotechnology, Sri Venkateswara University, Tirupati, AP-517502 India
| | | |
Collapse
|
50
|
Ouyang X, Feng Y, Cui C, Li Y, Zhang L, Wang H. Improving generalizability of drug-target binding prediction by pre-trained multi-view molecular representations. Bioinformatics 2024; 41:btaf002. [PMID: 39776159 PMCID: PMC11751634 DOI: 10.1093/bioinformatics/btaf002] [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: 06/25/2024] [Revised: 12/12/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
MOTIVATION Most drugs start on their journey inside the body by binding the right target proteins. This is the reason that numerous efforts have been devoted to predicting the drug-target binding during drug development. However, the inherent diversity among molecular properties, coupled with limited training data availability, poses challenges to the accuracy and generalizability of these methods beyond their training domain. RESULTS In this work, we proposed a neural networks construction for high accurate and generalizable drug-target binding prediction, named Pre-trained Multi-view Molecular Representations (PMMR). The method uses pre-trained models to transfer representations of target proteins and drugs to the domain of drug-target binding prediction, mitigating the issue of poor generalizability stemming from limited data. Then, two typical representations of drug molecules, Graphs and SMILES strings, are learned respectively by a Graph Neural Network and a Transformer to achieve complementarity between local and global features. PMMR was evaluated on drug-target affinity and interaction benchmark datasets, and it derived preponderant performance contrast to peer methods, especially generalizability in cold-start scenarios. Furthermore, our state-of-the-art method was indicated to have the potential for drug discovery by a case study of cyclin-dependent kinase 2. AVAILABILITY AND IMPLEMENTATION https://github.com/NENUBioCompute/PMMR.
Collapse
Affiliation(s)
- Xike Ouyang
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, Changchun, Jilin 130117, China
| | - Yannuo Feng
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, Changchun, Jilin 130117, China
| | - Chen Cui
- School of Computer Science and Engineering, Changchun University of Technology, Changchun, Jilin 130051, China
| | - Yunhe Li
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, Changchun, Jilin 130117, China
| | - Li Zhang
- School of Computer Science and Engineering, Changchun University of Technology, Changchun, Jilin 130051, China
| | - Han Wang
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, Changchun, Jilin 130117, China
| |
Collapse
|