1
|
Yadav MK, Dahiya V, Tripathi MK, Chaturvedi N, Rashmi M, Ghosh A, Raj VS. Unleashing the future: The revolutionary role of machine learning and artificial intelligence in drug discovery. Eur J Pharmacol 2024; 985:177103. [PMID: 39515559 DOI: 10.1016/j.ejphar.2024.177103] [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: 08/01/2024] [Revised: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Drug discovery is a complex and multifaceted process aimed at identifying new therapeutic compounds with the potential to treat various diseases. Traditional methods of drug discovery are often time-consuming, expensive, and characterized by low success rates. Because of this, there is an urgent need to improve the drug development process using new technologies. The integration of the current state-of-art of artificial intelligence (AI) and machine learning (ML) approaches with conventional methods will enhance the efficiency and effectiveness of pharmaceutical research. This review highlights the transformative impact of AI and ML in drug discovery, discussing current applications, challenges, and future directions in harnessing these technologies to accelerate the development of innovative therapeutics. We have discussed the latest developments in AI and ML technologies to streamline several stages of drug discovery, from target identification and validation to lead optimization and preclinical studies.
Collapse
Affiliation(s)
- Manoj Kumar Yadav
- Department of Biomedical Engineering, SRM University Delhi-NCR, Sonepat, Haryana, India.
| | - Vandana Dahiya
- Department of Biomedical Engineering, SRM University Delhi-NCR, Sonepat, Haryana, India
| | | | - Navaneet Chaturvedi
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Mayank Rashmi
- ICAR-Indian Agricultural Statistics Research Institute, New Delhi, India
| | - Arabinda Ghosh
- Department of Molecular Biology and Bioinformatics, Tripura University, Suryamaninagar, Tripura, India
| | - V Samuel Raj
- Center for Drug Design Discovery and Development (C4D), SRM University Delhi-NCR, Sonepat, Haryana, India.
| |
Collapse
|
2
|
Nangia AK. Molecular tweaking by generative cheminformatics and ligand-protein structures for rational drug discovery. Bioorg Chem 2024; 153:107920. [PMID: 39489080 DOI: 10.1016/j.bioorg.2024.107920] [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: 08/19/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
The purpose of this review is two-fold: (1) to summarize artificial intelligence and machine learning approaches and document the role of ligand-protein structures in directing drug discovery; (2) to present examples of drugs from the recent literature (past decade) of case studies where such strategies have been applied to accelerate the discovery pipeline. Compared to 50 years ago when drug discovery was largely a synthetic chemist driven research exercise, today a holistic approach needs to be adopted with seamless integration between synthetic and medicinal chemistry, supramolecular complexes, computations, artificial intelligence, machine learning, structural biology, chemical biology, diffraction analytical tools, drugs databases, and pharmacology. The urgency for an integrated and collaborative platform to accelerate drug discovery in an academic setting is emphasized.
Collapse
Affiliation(s)
- Ashwini K Nangia
- School of Chemistry, University of Hyderabad, Hyderabad 500 046, India.
| |
Collapse
|
3
|
Kint S, Dolfsma W, Robinson D. Strategic partnerships for AI-driven drug discovery: The role of relational dynamics. Drug Discov Today 2024; 29:104242. [PMID: 39547391 DOI: 10.1016/j.drudis.2024.104242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Artificial intelligence-driven drug discovery (AIDD) companies hold significant promise for transforming pharmaceutical development, yet little is known about how they manage partnerships with established pharmaceutical firms. To address this research gap, our study explores how AIDD companies develop and leverage relational capabilities to enhance collaboration effectiveness. Through a case study approach, we focus on four key relational aspects: identifying complementary capabilities, establishing effective governance mechanisms, creating relationship-specific assets, and developing interfirm knowledge-sharing routines. Our findings demonstrate that particularly effective governance of intellectual property is essential for partnership success. We offer actionable recommendations for AIDD companies to strengthen collaborations, thereby contributing to the realization of AI's potential in drug discovery.
Collapse
Affiliation(s)
- Stefan Kint
- Wageningen University & Research, Wageningen, the Netherlands
| | - Wilfred Dolfsma
- Wageningen University & Research, Wageningen, the Netherlands.
| | | |
Collapse
|
4
|
Tredup C, Ackloo S, Beck H, Brown PJ, Bullock AN, Ciulli A, Dikic I, Edfeldt K, Edwards AM, Elkins JM, Farin HF, Fon EA, Gstaiger M, Günther J, Gustavsson AL, Häberle S, Isigkeit L, Huber KVM, Kotschy A, Krämer O, Leach AR, Marsden BD, Matsui H, Merk D, Montel F, Mulder MPC, Müller S, Owen DR, Proschak E, Röhm S, Stolz A, Sundström M, von Delft F, Willson TM, Arrowsmith CH, Knapp S. Toward target 2035: EUbOPEN - a public-private partnership to enable & unlock biology in the open. RSC Med Chem 2024:d4md00735b. [PMID: 39618964 PMCID: PMC11605244 DOI: 10.1039/d4md00735b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024] Open
Abstract
Target 2035 is a global initiative that seeks to identify a pharmacological modulator of most human proteins by the year 2035. As part of an ongoing series of annual updates of this initiative, we summarise here the efforts of the EUbOPEN project whose objectives and results are making a strong contribution to the goals of Target 2035. EUbOPEN is a public-private partnership with four pillars of activity: (1) chemogenomic library collections, (2) chemical probe discovery and technology development for hit-to-lead chemistry, (3) profiling of bioactive compounds in patient-derived disease assays, and (4) collection, storage and dissemination of project-wide data and reagents. The substantial outputs of this programme include a chemogenomic compound library covering one third of the druggable proteome, as well as 100 chemical probes, both profiled in patient derived assays, as well as hundreds of data sets deposited in existing public data repositories and a project-specific data resource for exploring EUbOPEN outputs.
Collapse
Affiliation(s)
- Claudia Tredup
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| | - Suzanne Ackloo
- Structural Genomics Consortium, University of Toronto - St George Campus 101 College Street, MaRS Center South Tower 7th Floor Toronto Canada
| | - Hartmut Beck
- Drug Discovery Sciences, Research & Development, Pharmaceuticals, Bayer AG Wuppertal Nordrhein-Westfalen Germany
| | - Peter J Brown
- Structural Genomics Consortium, University of North Carolina at Chapel Hill Campus Box 7356, 120 Mason Farm Road, GMB 1070 Chapel Hill North Carolina USA
| | - Alex N Bullock
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
| | - Alessio Ciulli
- Centre for Targeted Protein Degradation, University of Dundee, School of Life Sciences 1 James Lindsay Place DD1 5JJ Dundee UK
| | - Ivan Dikic
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty Frankfurt am Main Germany
- Buchmann Institute for Molecular Lifesciences, Goethe University Frankfurt Frankfurt am Main Germany
| | - Kristina Edfeldt
- Structural Genomics Consortium, Department of Medicine, Karolinska University Hospital and Karolinska Institutet Stockholm Sweden
| | - Aled M Edwards
- Structural Genomics Consortium, University of Toronto - St George Campus 101 College Street, MaRS Center South Tower 7th Floor Toronto Canada
| | - Jonathan M Elkins
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
| | - Henner F Farin
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy Frankfurt am Main Hessen Germany
| | - Edward A Fon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital (The Neuro), McGill University Montreal Canada
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology ETH Zürich Zurich ZH Switzerland
| | | | - Anna-Lena Gustavsson
- Chemical Biology Consortium Sweden, Department of Medical Biochemistry & Biophysics, Karolinska Institute Stockholm Sweden
| | - Sandra Häberle
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| | - Laura Isigkeit
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
| | - Kilian V M Huber
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
| | - Andras Kotschy
- Servier Research Institute of Medicinal Chemistry Budapest Hungary
| | - Oliver Krämer
- Discovery Research Coordination, Boehringer Ingelheim International GmbH Binger Straße 173 55216 Ingelheim am Rhein Germany
| | - Andrew R Leach
- European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus Hinxton Cambridge UK
| | - Brian D Marsden
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
| | - Hisanori Matsui
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited Fujisawa Kanagawa Japan
| | - Daniel Merk
- Ludwig-Maximilians-Universitat Munchen Munchen Germany
| | - Florian Montel
- Discovery Research Coordination, Boehringer Ingelheim Pharma GmbH & Co. KG Birkendorfer Straße 65 88397 Biberach an der Riss Germany
| | - Monique P C Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center Leiden The Netherlands
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| | | | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP Theodor-Stern-Kai 7 60596 Frankfurt am Main Germany
| | - Sandra Röhm
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| | - Alexandra Stolz
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty Frankfurt am Main Germany
- Buchmann Institute for Molecular Lifesciences, Goethe University Frankfurt Frankfurt am Main Germany
| | - Michael Sundström
- Structural Genomics Consortium, Department of Medicine, Karolinska University Hospital and Karolinska Institutet Stockholm Sweden
| | - Frank von Delft
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
- Diamond Light Source, Harwell Science and Innovation Campus Didcot OX11 0DE UK
| | - Timothy M Willson
- Structural Genomics Consortium, University of North Carolina at Chapel Hill Campus Box 7356, 120 Mason Farm Road, GMB 1070 Chapel Hill North Carolina USA
| | - Cheryl H Arrowsmith
- Structural Genomics Consortium, University of Toronto - St George Campus 101 College Street, MaRS Center South Tower 7th Floor Toronto Canada
- Princess Margaret Cancer Centre Toronto Ontario M5G 1L7 Canada
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| |
Collapse
|
5
|
Luo Z, Wu W, Sun Q, Wang J. Accurate and transferable drug-target interaction prediction with DrugLAMP. Bioinformatics 2024; 40:btae693. [PMID: 39570605 PMCID: PMC11629708 DOI: 10.1093/bioinformatics/btae693] [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: 07/17/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024] Open
Abstract
MOTIVATION Accurate prediction of drug-target interactions (DTIs), especially for novel targets or drugs, is crucial for accelerating drug discovery. Recent advances in pretrained language models (PLMs) and multi-modal learning present new opportunities to enhance DTI prediction by leveraging vast unlabeled molecular data and integrating complementary information from multiple modalities. RESULTS We introduce DrugLAMP (PLM-assisted multi-modal prediction), a PLM-based multi-modal framework for accurate and transferable DTI prediction. DrugLAMP integrates molecular graph and protein sequence features extracted by PLMs and traditional feature extractors. We introduce two novel multi-modal fusion modules: (i) pocket-guided co-attention (PGCA), which uses protein pocket information to guide the attention mechanism on drug features, and (ii) paired multi-modal attention (PMMA), which enables effective cross-modal interactions between drug and protein features. These modules work together to enhance the model's ability to capture complex drug-protein interactions. Moreover, the contrastive compound-protein pre-training (2C2P) module enhances the model's generalization to real-world scenarios by aligning features across modalities and conditions. Comprehensive experiments demonstrate DrugLAMP's state-of-the-art performance on both standard benchmarks and challenging settings simulating real-world drug discovery, where test drugs/targets are unseen during training. Visualizations of attention maps and application to predict cryptic pockets and drug side effects further showcase DrugLAMP's strong interpretability and generalizability. Ablation studies confirm the contributions of the proposed modules. AVAILABILITY AND IMPLEMENTATION Source code and datasets are freely available at https://github.com/Lzcstan/DrugLAMP. All data originate from public sources.
Collapse
Affiliation(s)
- Zhengchao Luo
- Department of Big Data and Biomedical AI, College of Future Technology, Peking University, Beijing 100871, China
| | - Wei Wu
- Department of Big Data and Biomedical AI, College of Future Technology, Peking University, Beijing 100871, China
| | - Qichen Sun
- School of Mathematical Sciences, Peking University, Beijing 100871, China
| | - Jinzhuo Wang
- Department of Big Data and Biomedical AI, College of Future Technology, Peking University, Beijing 100871, China
| |
Collapse
|
6
|
Tiwari R, Dev D, Thalla M, Aher VD, Mundada AB, Mundada PA, Vaghela K. Nano-enabled pharmacogenomics: revolutionizing personalized drug therapy. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024:1-26. [PMID: 39589779 DOI: 10.1080/09205063.2024.2431426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/07/2024] [Indexed: 11/27/2024]
Abstract
The combination of pharmacogenomics and nanotechnology science of pharmacogenomics into a highly advanced single entity has given birth to personalized medicine known as nano-enabled pharmacogenomics. This review article covers all aspects starting from pharmacogenomics to gene editing tools, how these have evolved or are likely to be evolved for pharmacogenomic application, and how these can be delivered using nanoparticle delivery systems. In this prior work, we explore the evolution of pharmacogenomics over the years, as well as new achievements in the field of genomic sciences, the challenges in drug creation, and application of the strategy of personalized medicine. Particular attention is paid to how nanotechnology helps avoid the problems that accompanied the development of pharmacogenomics earlier, for example, the question of drug resistance and targeted delivery. We also review the latest developments in nano-enabled pharmacogenomics, such as the coupling with other nanobio-technologies, artificial intelligence, and machine learning in pharmacogenomics, and the ethical and regulatory aspects of these developing technologies. The possible uses of nanotechnology in improving the chances of pated and treating drug-resistant cancers are exemplified by case studies together with the current clinical uses of nanotechnology. In the last section, we discuss the future trends and research prospects in this dynamically growing area, stressing the importance of further advancements and collaborations which will advance the nano-enabled pharmacogenomics to their maximum potential.
Collapse
Affiliation(s)
- Ruchi Tiwari
- Psit-Pranveer Singh Institute of Technology (Pharmacy), Kanpur-Agra-Delhi National, Kanpur, India
| | - Dhruv Dev
- Department of Pharmacy, Shivalik College of Pharmacy Nangal, Rupnagar, India
| | - Maharshi Thalla
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, Kingsville, TX, USA
| | - Vaibhav Dagaji Aher
- Department of Pharmaceutical Medicine, Maharashtra University of Health Sciences, Nashik, India
| | - Anand Badrivishal Mundada
- Department of Pharmacy, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | | | - Krishna Vaghela
- Department of Pharmacy, Saraswati Institute of Pharmaceutical Sciences, National Forensic Sciences University, Gandhinagar, India
| |
Collapse
|
7
|
Son A, Park J, Kim W, Yoon Y, Lee S, Ji J, Kim H. Recent Advances in Omics, Computational Models, and Advanced Screening Methods for Drug Safety and Efficacy. TOXICS 2024; 12:822. [PMID: 39591001 PMCID: PMC11598288 DOI: 10.3390/toxics12110822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/10/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024]
Abstract
It is imperative to comprehend the mechanisms that underlie drug toxicity in order to enhance the efficacy and safety of novel therapeutic agents. The capacity to identify molecular pathways that contribute to drug-induced toxicity has been significantly enhanced by recent developments in omics technologies, such as transcriptomics, proteomics, and metabolomics. This has enabled the early identification of potential adverse effects. These insights are further enhanced by computational tools, including quantitative structure-activity relationship (QSAR) analyses and machine learning models, which accurately predict toxicity endpoints. Additionally, technologies such as physiologically based pharmacokinetic (PBPK) modeling and micro-physiological systems (MPS) provide more precise preclinical-to-clinical translation, thereby improving drug safety assessments. This review emphasizes the synergy between sophisticated screening technologies, in silico modeling, and omics data, emphasizing their roles in reducing late-stage drug development failures. Challenges persist in the integration of a variety of data types and the interpretation of intricate biological interactions, despite the progress that has been made. The development of standardized methodologies that further enhance predictive toxicology is contingent upon the ongoing collaboration between researchers, clinicians, and regulatory bodies. This collaboration ensures the development of therapeutic pharmaceuticals that are more effective and safer.
Collapse
Affiliation(s)
- Ahrum Son
- Department of Molecular Medicine, Scripps Research, San Diego, CA 92037, USA;
| | - Jongham Park
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (Y.Y.); (S.L.)
| | - Woojin Kim
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (Y.Y.); (S.L.)
| | - Yoonki Yoon
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (Y.Y.); (S.L.)
| | - Sangwoon Lee
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (Y.Y.); (S.L.)
| | - Jaeho Ji
- Department of Convergent Bioscience and Informatics, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea;
| | - Hyunsoo Kim
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (Y.Y.); (S.L.)
- Department of Convergent Bioscience and Informatics, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea;
- Protein AI Design Institute, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
- SCICS, Prove Beyond AI, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| |
Collapse
|
8
|
Kamuntavičius G, Prat A, Paquet T, Bastas O, Aty HA, Sun Q, Andersen CB, Harman J, Siladi ME, Rines DR, Flatters SJL, Tal R, Norvaišas P. Accelerated hit identification with target evaluation, deep learning and automated labs: prospective validation in IRAK1. J Cheminform 2024; 16:127. [PMID: 39543721 PMCID: PMC11566907 DOI: 10.1186/s13321-024-00914-0] [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: 12/03/2023] [Accepted: 10/10/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Target identification and hit identification can be transformed through the application of biomedical knowledge analysis, AI-driven virtual screening and robotic cloud lab systems. However there are few prospective studies that evaluate the efficacy of such integrated approaches. RESULTS We synergistically integrate our in-house-developed target evaluation (SpectraView) and deep-learning-driven virtual screening (HydraScreen) tools with an automated robotic cloud lab designed explicitly for ultra-high-throughput screening, enabling us to validate these platforms experimentally. By employing our target evaluation tool to select IRAK1 as the focal point of our investigation, we prospectively validate our structure-based deep learning model. We can identify 23.8% of all IRAK1 hits within the top 1% of ranked compounds. The model outperforms traditional virtual screening techniques and offers advanced features such as ligand pose confidence scoring. Simultaneously, we identify three potent (nanomolar) scaffolds from our compound library, 2 of which represent novel candidates for IRAK1 and hold promise for future development. CONCLUSION This study provides compelling evidence for SpectraView and HydraScreen to provide a significant acceleration in the processes of target identification and hit discovery. By leveraging Ro5's HydraScreen and Strateos' automated labs in hit identification for IRAK1, we show how AI-driven virtual screening with HydraScreen could offer high hit discovery rates and reduce experimental costs. SCIENTIFIC CONTRIBUTION We present an innovative platform that leverages Knowledge graph-based biomedical data analytics and AI-driven virtual screening integrated with robotic cloud labs. Through an unbiased, prospective evaluation we show the reliability and robustness of HydraScreen in virtual and high-throughput screening for hit identification in IRAK1. Our platforms and innovative tools can expedite the early stages of drug discovery.
Collapse
Affiliation(s)
| | - Alvaro Prat
- AI Chemistry, Ro5, 2801 Gateway Drive, Irving, 75063, TX, USA.
| | - Tanya Paquet
- AI Chemistry, Ro5, 2801 Gateway Drive, Irving, 75063, TX, USA
| | - Orestis Bastas
- AI Chemistry, Ro5, 2801 Gateway Drive, Irving, 75063, TX, USA
| | | | - Qing Sun
- Strateos, 3565 Haven Ave Suite 3, Menlo Park, 94025, CA, USA
| | | | - John Harman
- Strateos, 3565 Haven Ave Suite 3, Menlo Park, 94025, CA, USA
| | - Marc E Siladi
- Strateos, 3565 Haven Ave Suite 3, Menlo Park, 94025, CA, USA
| | - Daniel R Rines
- Strateos, 3565 Haven Ave Suite 3, Menlo Park, 94025, CA, USA
| | | | - Roy Tal
- AI Chemistry, Ro5, 2801 Gateway Drive, Irving, 75063, TX, USA.
| | | |
Collapse
|
9
|
Wang K, Zhu S, Zhang Y, Wang Y, Bian Z, Lu Y, Shao Q, Jin X, Xu X, Mo R. Targeting the GTPase RAN by liposome delivery for tackling cancer stemness-emanated therapeutic resistance. J Control Release 2024; 375:589-600. [PMID: 39245420 DOI: 10.1016/j.jconrel.2024.09.007] [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: 04/14/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Cancer therapeutic resistance as a common hallmark of cancer is often responsible for treatment failure and poor patient survival. Cancer stem-like cells (CSCs) are one of the main contributors to therapeutic resistance, cancer relapse and metastasis. Through screening from our in-house library of natural products, we found polyphyllin II (PPII) as a potent anti-CSC compound for triple-negative breast cancer (TNBC). To enhance anti-CSC selectivity and improve druggability of PPII, we leverage the liposome-mediated delivery technique for increasing solubility of PPII, and more significantly, attaining broader therapeutic window. Liposomal PPII demonstrates its marked potency to inhibit tumor growth, post-surgical recurrence and metastasis compared to commercial liposomal chemotherapeutics in the mouse models of CSC-enriched TNBC tumor. We further identify PPII as an inhibitor of the Ras-related nuclear (RAN) protein whose upregulated expression is correlated with poor clinical outcomes. The direct binding of PPII to RAN reduces TNBC stemness, thereby suppressing tumor progression. Our work offers a significance from drug discovery to drug delivery benefiting from liposome technique for targeted treatment of high-stemness tumor.
Collapse
Affiliation(s)
- Kaili Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Sitong Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Ying Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yuqian Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Zhenqian Bian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yougong Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Quanlin Shao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Xiang Jin
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310032, China
| | - Xiaojun Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; Department of Pharmacy, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Center for Innovative Traditional Chinese Medicine Target and New Drug Research, International Institutes of Medicine, Zhejiang University, Yiwu 322001, Zhejiang, China.
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
10
|
Shrinidhi A, Dwyer TS, Scott JA, Watts VJ, Flaherty DP. Pyrazolo-Pyrimidinones with Improved Solubility and Selective Inhibition of Adenylyl Cyclase Type 1 Activity for Treatment of Inflammatory Pain. J Med Chem 2024; 67:18290-18316. [PMID: 39404162 DOI: 10.1021/acs.jmedchem.4c01645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Adenylyl cyclase isoform 1 (AC1) is considered a promising target for treating inflammatory pain. Our group identified the pyrazolyl-pyrimidinone scaffold as potent and selective inhibitors of Ca2+/CaM-mediated AC1 activity; however, the molecules suffered from poor aqueous solubility. The current study presents a strategy to improve aqueous solubility of the scaffold by reduction of crystal packing energy and increasing rotational degrees of freedom within the molecule. Structure-activity and property relationship studies identified the second generation lead 7-47A (AC10142A) that demonstrated and AC1 IC50 value of 0.26 μM and aqueous solubility of 74 ± 7 μM. After in vitro ADME characterization, the scaffold advanced to in vivo pharmacokinetic evaluation, demonstrating adequate levels of exposure. Finally, 7-47A exhibited antiallodynic efficacy in a rat complete Freund's adjuvant model for inflammatory pain showing improvement over previous iterations of this scaffold. These results further validate AC1 inhibition as a viable therapeutic strategy for treating chronic and inflammatory pain.
Collapse
Affiliation(s)
- Annadka Shrinidhi
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Tiffany S Dwyer
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jason A Scott
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Val J Watts
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Drug Discovery, West Lafayette, Indiana 47907, United States
- Purdue Institute for Integrative Neuroscience, 207 South Martin Jischke Dr., West Lafayette, Indiana 47907, United States
| | - Daniel P Flaherty
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Drug Discovery, West Lafayette, Indiana 47907, United States
- Purdue Institute for Integrative Neuroscience, 207 South Martin Jischke Dr., West Lafayette, Indiana 47907, United States
| |
Collapse
|
11
|
Welsch M, Hirte S, Kirchmair J. Deciphering Molecular Embeddings with Centered Kernel Alignment. J Chem Inf Model 2024; 64:7303-7312. [PMID: 39321215 PMCID: PMC11482110 DOI: 10.1021/acs.jcim.4c00837] [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: 05/14/2024] [Revised: 07/24/2024] [Accepted: 09/06/2024] [Indexed: 09/27/2024]
Abstract
Analyzing machine learning models, especially nonlinear ones, poses significant challenges. In this context, centered kernel alignment (CKA) has emerged as a promising model analysis tool that assesses the similarity between two embeddings. CKA's efficacy depends on selecting a kernel that adequately captures the underlying properties of the compared models. The model analysis tool was designed for neural networks (NNs) with their invariance to data rotation in mind and has been successfully employed in various scientific domains. However, CKA has rarely been adopted in cheminformatics, partly because of the popularity of the random forest (RF) machine learning algorithm, which is not rotationally invariant. In this work, we present the adaptation of CKA that builds on the RF kernel to match the properties of RF. As part of the method validation, we show that the model analysis method is well-correlated with the prediction similarity of RF models. Furthermore, we demonstrate how CKA with the RF kernel can be utilized to analyze and explain the behavior of RF models derived from molecular and rooted fingerprints.
Collapse
Affiliation(s)
- Matthias Welsch
- Department of Pharmaceutical Sciences, Division of
Pharmaceutical Chemistry, Faculty of Life Sciences, University of
Vienna, Josef-Holaubek-Platz 2, Vienna 1090,
Austria
- Christian Doppler Laboratory for Molecular Informatics
in the Biosciences, Department for Pharmaceutical Sciences, University of
Vienna, Vienna 1090, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional
and Sport Sciences (PhaNuSpo), University of Vienna, Vienna
1090, Austria
| | - Steffen Hirte
- Department of Pharmaceutical Sciences, Division of
Pharmaceutical Chemistry, Faculty of Life Sciences, University of
Vienna, Josef-Holaubek-Platz 2, Vienna 1090,
Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional
and Sport Sciences (PhaNuSpo), University of Vienna, Vienna
1090, Austria
| | - Johannes Kirchmair
- Department of Pharmaceutical Sciences, Division of
Pharmaceutical Chemistry, Faculty of Life Sciences, University of
Vienna, Josef-Holaubek-Platz 2, Vienna 1090,
Austria
- Christian Doppler Laboratory for Molecular Informatics
in the Biosciences, Department for Pharmaceutical Sciences, University of
Vienna, Vienna 1090, Austria
| |
Collapse
|
12
|
Allemailem KS, Almatroudi A, Alrumaihi F, Alradhi AE, Theyab A, Algahtani M, Alhawas MO, Dobie G, Moawad AA, Rahmani AH, Khan AA. Current Updates of CRISPR/Cas System and Anti-CRISPR Proteins: Innovative Applications to Improve the Genome Editing Strategies. Int J Nanomedicine 2024; 19:10185-10212. [PMID: 39399829 PMCID: PMC11471075 DOI: 10.2147/ijn.s479068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024] Open
Abstract
The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated sequence (CRISPR/Cas) system is a cutting-edge genome-editing tool employed to explore the functions of normal and disease-related genes. The CRISPR/Cas system has a remarkable diversity in the composition and architecture of genomic loci and Cas protein sequences. Owing to its excellent efficiency and specificity, this system adds an outstanding dimension to biomedical research on genetic manipulation of eukaryotic cells. However, safe, efficient, and specific delivery of this system to target cells and tissues and their off-target effects are considered critical bottlenecks for the therapeutic applications. Recently discovered anti-CRISPR proteins (Acr) play a significant role in limiting the effects of this system. Acrs are relatively small proteins that are highly specific to CRISPR variants and exhibit remarkable structural diversity. The in silico approaches, crystallography, and cryo-electron microscopy play significant roles in elucidating the mechanisms of action of Acrs. Acrs block the CRISPR/Cas system mainly by employing four mechanisms: CRISPR/Cas complex assembly interruption, target-binding interference, target cleavage prevention, and degradation of cyclic oligonucleotide signaling molecules. Engineered CRISPR/Cas systems are frequently used in gene therapy, diagnostics, and functional genomics. Understanding the molecular mechanisms underlying Acr action may help in the safe and effective use of CRISPR/Cas tools for genetic modification, particularly in the context of medicine. Thus, attempts to regulate prokaryotic CRISPR/Cas surveillance complexes will advance the development of antimicrobial drugs and treatment of human diseases. In this review, recent updates on CRISPR/Cas systems, especially CRISPR/Cas9 and Acrs, and their novel mechanistic insights are elaborated. In addition, the role of Acrs in the novel applications of CRISPP/Cas biotechnology for precise genome editing and other applications is discussed.
Collapse
Affiliation(s)
- Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Arwa Essa Alradhi
- General Administration for Infectious Disease Control, Ministry of Health, Riyadh 12382, Saudi Arabia
| | - Abdulrahman Theyab
- Department of Laboratory & Blood Bank, Security Forces Hospital, Mecca 21955, Saudi Arabia
- College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | - Mohammad Algahtani
- Department of Laboratory & Blood Bank, Security Forces Hospital, Mecca 21955, Saudi Arabia
| | | | - Gasim Dobie
- Department of Medical Laboratory Technology, College of Nursing and Health Sciences, Jazan University, Gizan, 82911, Saudi Arabia
| | - Amira A Moawad
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Jena 07743, Germany
- Animal Health Research Institute, Agriculture Research Center, Giza 12618, Egypt
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
13
|
Morvan J, Kuijpers KPL, Fanfair D, Tang B, Bartkowiak K, van Eynde L, Renders E, Alcazar J, Buijnsters PJJA, Carvalho MA, Jones AX. Electrochemical C-O and C-N Arylation using Alternating Polarity in flow for Compound Libraries. Angew Chem Int Ed Engl 2024:e202413383. [PMID: 39383014 DOI: 10.1002/anie.202413383] [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: 07/16/2024] [Revised: 09/06/2024] [Accepted: 10/07/2024] [Indexed: 10/11/2024]
Abstract
Etherification and amination of aryl halide scaffolds are commonly used reactions in parallel medicinal chemistry to rapidly scan structure-activity relationships with abundant building blocks. Electrochemical methods for aryl etherification and amination demonstrate broad functional group tolerance and extended nucleophile scope compared to traditional methods. Nevertheless, there is a need for robust and scale-transferable workflows for electrochemical compound library synthesis. Herein we describe a platform for automated electrochemical synthesis of C-X arylation (X=NH, OH) in flow to access compound libraries. A comprehensive Design of Experiment (DoE) study identifies an optimal protocol which generates high yields across>30 aryl halide scaffolds, diverse amines (including electron-deficient sulfonamides, sulfoximines, amides, and anilines) and alcohols (including serine residues within peptides). Reaction sequences are automated on commercially available equipment to generate libraries of anilines and aryl ethers. The unprecedented application of potentiostatic alternating polarity in flow is essential to avoid accumulating electrode passivation. Moreover, it enables reactions to be performed in air, without supporting electrolyte and with high reproducibility over consecutive runs. Our method represents a powerful means to rapidly generate nucleophile independent C-X arylation compound libraries using flow electrochemistry.
Collapse
Affiliation(s)
- Jennifer Morvan
- Global Discovery Chemistry, Janssen Research and Development, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Koen P L Kuijpers
- API SM Technology, Janssen Research and Development, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Dayne Fanfair
- API SM Technology, Janssen Research and Development, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Bingqing Tang
- Global Discovery Chemistry, Janssen Research and Development, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Karolina Bartkowiak
- Global Discovery Chemistry, Janssen Research and Development, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Lars van Eynde
- Global Discovery Chemistry, Janssen Research and Development, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Evelien Renders
- Global Discovery Chemistry, Janssen Research and Development, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Jesus Alcazar
- Chemical Capabilities, Analytical & Purification, Global Discovery Chemistry, Janssen-Cilag, S.A., C/Jarama 75, 45007, Toledo, Spain
| | - Peter J J A Buijnsters
- Global Discovery Chemistry, Janssen Research and Development, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Mary-Ambre Carvalho
- Global Discovery Chemistry, Janssen Research and Development, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Alexander X Jones
- Global Discovery Chemistry, Janssen Research and Development, Turnhoutseweg 30, 2340, Beerse, Belgium
| |
Collapse
|
14
|
Doyle SE, Cazzola CN, Coleman CM. Design considerations when creating a high throughput screen-compatible in vitro model of osteogenesis. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100184. [PMID: 39313131 DOI: 10.1016/j.slasd.2024.100184] [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: 06/07/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
Inducing osteogenic differentiation in vitro is useful for the identification and development of bone regeneration therapies as well as modelling bone disorders. To couple in vitro models with high throughput screening techniques retains the assay's relevance in research while increasing its therapeutic impact. Miniaturizing, automating and/or digitalizing in vitro assays will reduce the required quantity of cells, biologic stimulants, culture/output assay reagents, time and cost. This review highlights the design and workflow considerations for creating a high throughput screen-compatible model of osteogenesis, comparing and contrasting osteogenic cell type, assay fabrication and culture methodology, osteogenic induction approach and repurposing existing output techniques.
Collapse
Affiliation(s)
- Stephanie E Doyle
- Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Science, University of Galway, Galway City, County Galway H91 FD82, Ireland.
| | - Courtney N Cazzola
- Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Science, University of Galway, Galway City, County Galway H91 FD82, Ireland
| | - Cynthia M Coleman
- Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Science, University of Galway, Galway City, County Galway H91 FD82, Ireland
| |
Collapse
|
15
|
Liu Y, Roccapriore K, Checa M, Valleti SM, Yang JC, Jesse S, Vasudevan RK. AEcroscopy: A Software-Hardware Framework Empowering Microscopy Toward Automated and Autonomous Experimentation. SMALL METHODS 2024; 8:e2301740. [PMID: 38639016 DOI: 10.1002/smtd.202301740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/31/2024] [Indexed: 04/20/2024]
Abstract
Microscopy has been pivotal in improving the understanding of structure-function relationships at the nanoscale and is by now ubiquitous in most characterization labs. However, traditional microscopy operations are still limited largely by a human-centric click-and-go paradigm utilizing vendor-provided software, which limits the scope, utility, efficiency, effectiveness, and at times reproducibility of microscopy experiments. Here, a coupled software-hardware platform is developed that consists of a software package termed AEcroscopy (short for Automated Experiments in Microscopy), along with a field-programmable-gate-array device with LabView-built customized acquisition scripts, which overcome these limitations and provide the necessary abstractions toward full automation of microscopy platforms. The platform works across multiple vendor devices on scanning probe microscopes and electron microscopes. It enables customized scan trajectories, processing functions that can be triggered locally or remotely on processing servers, user-defined excitation waveforms, standardization of data models, and completely seamless operation through simple Python commands to enable a plethora of microscopy experiments to be performed in a reproducible, automated manner. This platform can be readily coupled with existing machine-learning libraries and simulations, to provide automated decision-making and active theory-experiment optimization to turn microscopes from characterization tools to instruments capable of autonomous model refinement and physics discovery.
Collapse
Affiliation(s)
- Yongtao Liu
- Center for Nanophase Materials Sciences, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA
| | - Kevin Roccapriore
- Center for Nanophase Materials Sciences, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA
| | - Marti Checa
- Center for Nanophase Materials Sciences, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA
| | - Sai Mani Valleti
- Bredesen Center for Interdisciplinary Research, University of Tennessee, Knoxville, TN, 37996, USA
| | - Jan-Chi Yang
- Department of Physics, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Stephen Jesse
- Center for Nanophase Materials Sciences, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA
| | - Rama K Vasudevan
- Center for Nanophase Materials Sciences, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA
| |
Collapse
|
16
|
Milon TI, Wang Y, Fontenot RL, Khajouie P, Villinger F, Raghavan V, Xu W. Development of a novel representation of drug 3D structures and enhancement of the TSR-based method for probing drug and target interactions. Comput Biol Chem 2024; 112:108117. [PMID: 38852360 PMCID: PMC11390338 DOI: 10.1016/j.compbiolchem.2024.108117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/13/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Understanding the mechanisms underlying interactions between drugs and target proteins is critical for drug discovery. In our earlier studies, we introduced the Triangular Spatial Relationship (TSR)-based algorithm, which enables the representation of a protein's 3D structure as a vector of integers (TSR keys). These TSR keys correspond to substructures of the 3D structure of a protein and are computed based on the triangles constructed by all possible triples of Cα atoms within the protein. In this study, we report on a new TSR-based algorithm for probing drug and target interactions. Specifically, we have extended the previous algorithm in three novel directions: TSR keys for representing the 3D structure of a drug or a ligand, cross TSR keys between drugs and their targets and intra-residual TSR keys for phosphorylated amino acids. The outcomes illustrate the key contributions as follows: (i) The TSR-based method, which uses the TSR keys as features, is unique in its capability to interpret hierarchical relationships of drugs as well as drug - target complexes using common and specific TSR keys. (ii) The method can distinguish not only the binding sites from the rest of the protein structures, but also the binding sites of primary targets from those of off-targets. (iii) The method has the potential to correlate the 3D structures of drugs with their functions. (iv) Representation of 3D structures by TSR keys has its unique advantage in terms of ease of making searching for similar substructures across structure datasets easier. In summary, this study presents a novel computational methodology, with significant advantages, for providing insights into the mechanism underlying drug and target interactions.
Collapse
Affiliation(s)
- Tarikul I Milon
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA
| | - Yuhong Wang
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Ryan L Fontenot
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA
| | - Poorya Khajouie
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA; The Center for Advanced Computer Studies, University of Louisiana at Lafayette, LA 70504, USA
| | - Francois Villinger
- Department of Biology, University of Louisiana at Lafayette, New Iberia, LA 70560, USA
| | - Vijay Raghavan
- The Center for Advanced Computer Studies, University of Louisiana at Lafayette, LA 70504, USA
| | - Wu Xu
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA.
| |
Collapse
|
17
|
Ashraf SN, Blackwell JH, Holdgate GA, Lucas SCC, Solovyeva A, Storer RI, Whitehurst BC. Hit me with your best shot: Integrated hit discovery for the next generation of drug targets. Drug Discov Today 2024; 29:104143. [PMID: 39173704 DOI: 10.1016/j.drudis.2024.104143] [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: 05/31/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
Identification of high-quality hit chemical matter is of vital importance to the success of drug discovery campaigns. However, this goal is becoming ever harder to achieve as the targets entering the portfolios of pharmaceutical and biotechnology companies are increasingly trending towards novel and traditionally challenging to drug. This demand has fuelled the development and adoption of numerous new screening approaches, whereby the contemporary hit identification toolbox comprises a growing number of orthogonal and complementary technologies including high-throughput screening, fragment-based ligand design, affinity screening (affinity-selection mass spectrometry, differential scanning fluorimetry, DNA-encoded library screening), as well as increasingly sophisticated computational predictive approaches. Herein we describe how an integrated strategy for hit discovery, whereby multiple hit identification techniques are tactically applied, selected in the context of target suitability and resource priority, represents an optimal and often essential approach to maximise the likelihood of identifying quality starting points from which to develop the next generation of medicines.
Collapse
Affiliation(s)
- S Neha Ashraf
- Hit Discovery, Discovery Science, AstraZeneca R&D, Cambridge CB2 0AA, UK
| | - J Henry Blackwell
- Hit Discovery, Discovery Science, AstraZeneca R&D, Cambridge CB2 0AA, UK
| | | | - Simon C C Lucas
- Hit Discovery, Discovery Science, AstraZeneca R&D, Cambridge CB2 0AA, UK
| | - Alisa Solovyeva
- Hit Discovery, Discovery Science, AstraZeneca R&D, Gothenburg SE-431 83, Sweden
| | - R Ian Storer
- Hit Discovery, Discovery Science, AstraZeneca R&D, Cambridge CB2 0AA, UK.
| | | |
Collapse
|
18
|
Bharadwaj S, Deepika K, Kumar A, Jaiswal S, Miglani S, Singh D, Fartyal P, Kumar R, Singh S, Singh MP, Gaidhane AM, Kumar B, Jha V. Exploring the Artificial Intelligence and Its Impact in Pharmaceutical Sciences: Insights Toward the Horizons Where Technology Meets Tradition. Chem Biol Drug Des 2024; 104:e14639. [PMID: 39396920 DOI: 10.1111/cbdd.14639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/03/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024]
Abstract
The technological revolutions in computers and the advancement of high-throughput screening technologies have driven the application of artificial intelligence (AI) for faster discovery of drug molecules with more efficiency, and cost-friendly finding of hit or lead molecules. The ability of software and network frameworks to interpret molecular structures' representations and establish relationships/correlations has enabled various research teams to develop numerous AI platforms for identifying new lead molecules or discovering new targets for already established drug molecules. The prediction of biological activity, ADME properties, and toxicity parameters in early stages have reduced the chances of failure and associated costs in later clinical stages, which was observed at a high rate in the tedious, expensive, and laborious drug discovery process. This review focuses on the different AI and machine learning (ML) techniques with their applications mainly focused on the pharmaceutical industry. The applications of AI frameworks in the identification of molecular target, hit identification/hit-to-lead optimization, analyzing drug-receptor interactions, drug repurposing, polypharmacology, synthetic accessibility, clinical trial design, and pharmaceutical developments are discussed in detail. We have also compiled the details of various startups in AI in this field. This review will provide a comprehensive analysis and outline various state-of-the-art AI/ML techniques to the readers with their framework applications. This review also highlights the challenges in this field, which need to be addressed for further success in pharmaceutical applications.
Collapse
Affiliation(s)
- Shruti Bharadwaj
- Center for SeNSE, Indian Institute of Technology Delhi (IIT), New Delhi, India
| | - Kumari Deepika
- Department of Computer Engineering, Pune Institute of Computer Technology, Pune, India
| | - Asim Kumar
- Amity Institute of Pharmacy (AIP), Amity University Haryana, Manesar, India
| | - Shivani Jaiswal
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Shaweta Miglani
- Department of Education, Central University of Punjab, Bathinda, India
| | - Damini Singh
- IES Institute of Pharmacy, IES University, Bhopal, Madhya Pradesh, India
| | - Prachi Fartyal
- Department of Mathematics, Govt PG College Bajpur (US Nagar), Bazpur, Uttarakhand, India
| | - Roshan Kumar
- Department of Microbiology, Graphic Era (Deemed to be University), Dehradun, India
- Department of Microbiology, Central University of Punjab, VPO-Ghudda, Punjab, India
| | - Shareen Singh
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Mahendra Pratap Singh
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Abhay M Gaidhane
- Jawaharlal Nehru Medical College, and Global Health Academy, School of Epidemiology and Public Health, Datta Meghe Institute of Higher Education, Wardha, India
| | - Bhupinder Kumar
- Department of Pharmaceutical Science, Hemvati Nandan Bahuguna Garhwal (A Central) University, Srinagar, Uttarakhand, India
| | - Vibhu Jha
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
19
|
Filiz Y, Esposito A, De Maria C, Vozzi G, Yesil-Celiktas O. A comprehensive review on organ-on-chips as powerful preclinical models to study tissue barriers. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2024; 6:042001. [PMID: 39655848 DOI: 10.1088/2516-1091/ad776c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 09/04/2024] [Indexed: 12/18/2024]
Abstract
In the preclinical stage of drug development, 2D and 3D cell cultures under static conditions followed by animal models are utilized. However, these models are insufficient to recapitulate the complexity of human physiology. With the developing organ-on-chip (OoC) technology in recent years, human physiology and pathophysiology can be modeled better than traditional models. In this review, the need for OoC platforms is discussed and evaluated from both biological and engineering perspectives. The cellular and extracellular matrix components are discussed from a biological perspective, whereas the technical aspects such as the intricate working principles of these systems, the pivotal role played by flow dynamics and sensor integration within OoCs are elucidated from an engineering perspective. Combining these two perspectives, bioengineering applications are critically discussed with a focus on tissue barriers such as blood-brain barrier, ocular barrier, nasal barrier, pulmonary barrier and gastrointestinal barrier, featuring recent examples from the literature. Furthermore, this review offers insights into the practical utility of OoC platforms for modeling tissue barriers, showcasing their potential and drawbacks while providing future projections for innovative technologies.
Collapse
Affiliation(s)
- Yagmur Filiz
- Department of Development and Regeneration, Faculty of Medicine, KU Leuven, 8500 Kortrijk, Belgium
| | - Alessio Esposito
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Carmelo De Maria
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Giovanni Vozzi
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Ozlem Yesil-Celiktas
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Izmir, Turkey
- EgeSAM-Ege University Translational Pulmonary Research Center, Bornova, Izmir, Turkey
- ODTÜ MEMS Center, Ankara, Turkey
| |
Collapse
|
20
|
Sobhani N, D'Angelo A, Pittacolo M, Mondani G, Generali D. Future AI Will Most Likely Predict Antibody-Drug Conjugate Response in Oncology: A Review and Expert Opinion. Cancers (Basel) 2024; 16:3089. [PMID: 39272947 PMCID: PMC11394064 DOI: 10.3390/cancers16173089] [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: 07/31/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
The medical research field has been tremendously galvanized to improve the prediction of therapy efficacy by the revolution in artificial intelligence (AI). An earnest desire to find better ways to predict the effectiveness of therapy with the use of AI has propelled the evolution of new models in which it can become more applicable in clinical settings such as breast cancer detection. However, in some instances, the U.S. Food and Drug Administration was obliged to back some previously approved inaccurate models for AI-based prognostic models because they eventually produce inaccurate prognoses for specific patients who might be at risk of heart failure. In light of instances in which the medical research community has often evolved some unrealistic expectations regarding the advances in AI and its potential use for medical purposes, implementing standard procedures for AI-based cancer models is critical. Specifically, models would have to meet some general parameters for standardization, transparency of their logistic modules, and avoidance of algorithm biases. In this review, we summarize the current knowledge about AI-based prognostic methods and describe how they may be used in the future for predicting antibody-drug conjugate efficacy in cancer patients. We also summarize the findings of recent late-phase clinical trials using these conjugates for cancer therapy.
Collapse
Affiliation(s)
- Navid Sobhani
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alberto D'Angelo
- Department of Medicine, Northern General Hospital, Sheffield S5 7AT, UK
| | - Matteo Pittacolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
| | - Giuseppina Mondani
- Royal Infirmary Hospital, Foresterhill Health Campus, Aberdeen AB25 2ZN, UK
| | - Daniele Generali
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34100 Trieste, Italy
| |
Collapse
|
21
|
Dortaj H, Amani AM, Tayebi L, Azarpira N, Ghasemi Toudeshkchouei M, Hassanpour-Dehnavi A, Karami N, Abbasi M, Najafian-Najafabadi A, Zarei Behjani Z, Vaez A. Droplet-based microfluidics: an efficient high-throughput portable system for cell encapsulation. J Microencapsul 2024; 41:479-501. [PMID: 39077800 DOI: 10.1080/02652048.2024.2382744] [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: 12/01/2023] [Accepted: 07/17/2024] [Indexed: 07/31/2024]
Abstract
One of the goals of tissue engineering and regenerative medicine is restoring primary living tissue function by manufacturing a 3D microenvironment. One of the main challenges is protecting implanted non-autologous cells or tissues from the host immune system. Cell encapsulation has emerged as a promising technique for this purpose. It involves entrapping cells in biocompatible and semi-permeable microcarriers made from natural or synthetic polymers that regulate the release of cellular secretions. In recent years, droplet-based microfluidic systems have emerged as powerful tools for cell encapsulation in tissue engineering and regenerative medicine. These systems offer precise control over droplet size, composition, and functionality, allowing for creating of microenvironments that closely mimic native tissue. Droplet-based microfluidic systems have extensive applications in biotechnology, medical diagnosis, and drug discovery. This review summarises the recent developments in droplet-based microfluidic systems and cell encapsulation techniques, as well as their applications, advantages, and challenges in biology and medicine. The integration of these technologies has the potential to revolutionise tissue engineering and regenerative medicine by providing a precise and controlled microenvironment for cell growth and differentiation. By overcoming the immune system's challenges and enabling the release of cellular secretions, these technologies hold great promise for the future of regenerative medicine.
Collapse
Affiliation(s)
- Hengameh Dortaj
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, USA
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Ashraf Hassanpour-Dehnavi
- Tissue Engineering Lab, Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda Karami
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Abbasi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Atefeh Najafian-Najafabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zeinab Zarei Behjani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
22
|
Tom G, Schmid SP, Baird SG, Cao Y, Darvish K, Hao H, Lo S, Pablo-García S, Rajaonson EM, Skreta M, Yoshikawa N, Corapi S, Akkoc GD, Strieth-Kalthoff F, Seifrid M, Aspuru-Guzik A. Self-Driving Laboratories for Chemistry and Materials Science. Chem Rev 2024; 124:9633-9732. [PMID: 39137296 PMCID: PMC11363023 DOI: 10.1021/acs.chemrev.4c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Self-driving laboratories (SDLs) promise an accelerated application of the scientific method. Through the automation of experimental workflows, along with autonomous experimental planning, SDLs hold the potential to greatly accelerate research in chemistry and materials discovery. This review provides an in-depth analysis of the state-of-the-art in SDL technology, its applications across various scientific disciplines, and the potential implications for research and industry. This review additionally provides an overview of the enabling technologies for SDLs, including their hardware, software, and integration with laboratory infrastructure. Most importantly, this review explores the diverse range of scientific domains where SDLs have made significant contributions, from drug discovery and materials science to genomics and chemistry. We provide a comprehensive review of existing real-world examples of SDLs, their different levels of automation, and the challenges and limitations associated with each domain.
Collapse
Affiliation(s)
- Gary Tom
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Vector Institute
for Artificial Intelligence, 661 University Ave Suite 710, Toronto, Ontario M5G 1M1, Canada
| | - Stefan P. Schmid
- Department
of Chemistry and Applied Biosciences, ETH
Zurich, Vladimir-Prelog-Weg 1, CH-8093 Zurich, Switzerland
| | - Sterling G. Baird
- Acceleration
Consortium, 80 St. George
St, Toronto, Ontario M5S 3H6, Canada
| | - Yang Cao
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Acceleration
Consortium, 80 St. George
St, Toronto, Ontario M5S 3H6, Canada
| | - Kourosh Darvish
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Vector Institute
for Artificial Intelligence, 661 University Ave Suite 710, Toronto, Ontario M5G 1M1, Canada
- Acceleration
Consortium, 80 St. George
St, Toronto, Ontario M5S 3H6, Canada
| | - Han Hao
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Acceleration
Consortium, 80 St. George
St, Toronto, Ontario M5S 3H6, Canada
| | - Stanley Lo
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
| | - Sergio Pablo-García
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
| | - Ella M. Rajaonson
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Vector Institute
for Artificial Intelligence, 661 University Ave Suite 710, Toronto, Ontario M5G 1M1, Canada
| | - Marta Skreta
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Vector Institute
for Artificial Intelligence, 661 University Ave Suite 710, Toronto, Ontario M5G 1M1, Canada
| | - Naruki Yoshikawa
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Vector Institute
for Artificial Intelligence, 661 University Ave Suite 710, Toronto, Ontario M5G 1M1, Canada
| | - Samantha Corapi
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
| | - Gun Deniz Akkoc
- Forschungszentrum
Jülich GmbH, Helmholtz Institute
for Renewable Energy Erlangen-Nürnberg, Cauerstr. 1, 91058 Erlangen, Germany
- Department
of Chemical and Biological Engineering, Friedrich-Alexander Universität Erlangen-Nürnberg, Egerlandstr. 3, 91058 Erlangen, Germany
| | - Felix Strieth-Kalthoff
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- School of
Mathematics and Natural Sciences, University
of Wuppertal, Gaußstraße
20, 42119 Wuppertal, Germany
| | - Martin Seifrid
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Department
of Materials Science and Engineering, North
Carolina State University, Raleigh, North Carolina 27695, United States of America
| | - Alán Aspuru-Guzik
- Department
of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario M5S 3H6, Canada
- Department
of Computer Science, University of Toronto, 40 St. George St, Toronto, Ontario M5S 2E4, Canada
- Vector Institute
for Artificial Intelligence, 661 University Ave Suite 710, Toronto, Ontario M5G 1M1, Canada
- Acceleration
Consortium, 80 St. George
St, Toronto, Ontario M5S 3H6, Canada
- Department
of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
- Department
of Materials Science & Engineering, University of Toronto, Toronto, Ontario M5S 3E4, Canada
- Lebovic
Fellow, Canadian Institute for Advanced
Research (CIFAR), 661
University Ave, Toronto, Ontario M5G 1M1, Canada
| |
Collapse
|
23
|
Fralish Z, Reker D. Finding the most potent compounds using active learning on molecular pairs. Beilstein J Org Chem 2024; 20:2152-2162. [PMID: 39224230 PMCID: PMC11368049 DOI: 10.3762/bjoc.20.185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Active learning allows algorithms to steer iterative experimentation to accelerate and de-risk molecular optimizations, but actively trained models might still exhibit poor performance during early project stages where the training data is limited and model exploitation might lead to analog identification with limited scaffold diversity. Here, we present ActiveDelta, an adaptive approach that leverages paired molecular representations to predict improvements from the current best training compound to prioritize further data acquisition. We apply the ActiveDelta concept to both graph-based deep (Chemprop) and tree-based (XGBoost) models during exploitative active learning for 99 Ki benchmarking datasets. We show that both ActiveDelta implementations excel at identifying more potent inhibitors compared to the standard exploitative active learning implementations of Chemprop, XGBoost, and Random Forest. The ActiveDelta approach is also able to identify more chemically diverse inhibitors in terms of their Murcko scaffolds. Finally, deep models such as Chemprop trained on data selected through ActiveDelta approaches can more accurately identify inhibitors in test data created through simulated time-splits. Overall, this study highlights the large potential for molecular pairing approaches to further improve popular active learning strategies in low data regimes by enabling faster and more accurate identification of more diverse molecular hits against critical drug targets.
Collapse
Affiliation(s)
- Zachary Fralish
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Daniel Reker
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
24
|
Ai Z, Zhang L, Chen Y, Meng Y, Long Y, Xiao J, Yang Y, Guo W, Wang Y, Jiang J. Customizable Colorimetric Sensor Array via a High-Throughput Robot for Mitigation of Humidity Interference in Gas Sensing. ACS Sens 2024; 9:4143-4153. [PMID: 39086324 DOI: 10.1021/acssensors.4c01083] [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: 08/02/2024]
Abstract
One challenge for gas sensors is humidity interference, as dynamic humidity conditions can cause unpredictable fluctuations in the response signal to analytes, increasing quantitative detection errors. Here, we introduce a concept: Select humidity sensors from a pool to compensate for the humidity signal for each gas sensor. In contrast to traditional methods that extremely suppress the humidity response, the sensor pool allows for more accurate gas quantification across a broader range of application scenarios by supplying customized, high-dimensional humidity response data as extrinsic compensation. As a proof-of-concept, mitigation of humidity interference in colorimetric gas quantification was achieved in three steps. First, across a ten-dimensional variable space, an algorithm-driven high-throughput experimental robot discovered multiple local optimum regions where colorimetric humidity sensing formulations exhibited high evaluations on sensitivity, reversibility, response time, and color change extent for 10-90% relative humidity (RH) in room temperature (25 °C). Second, from the local optimum regions, 91 sensing formulations with diverse variables were selected to construct a parent colorimetric humidity sensor array as the sensor pool for humidity signal compensation. Third, the quasi-optimal sensor subarrays were identified as customized humidity signal compensation solutions for different gas sensing scenarios across an approximately full dynamic range of humidity (10-90% RH) using an ingenious combination optimization strategy, and two accurate quantitative detections were attained: one with a mean absolute percentage error (MAPE) reduction from 4.4 to 0.75% and the other from 5.48 to 1.37%. Moreover, the parent sensor array's excellent humidity selectivity was validated against 10 gases. This work demonstrates the feasibility and superiority of robot-assisted construction of a customizable parent colorimetric sensor array to mitigate humidity interference in gas quantification.
Collapse
Affiliation(s)
- Zhehong Ai
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
- Research Center for High Efficiency Computing System, Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
| | - Longhan Zhang
- Research Center for New Materials Computing, Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
- Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511458, China
| | - Yangguan Chen
- Research Center for New Materials Computing, Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
| | - Yu Meng
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing 100091, China
| | - Yifan Long
- Research Center for Space Computing System, Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
| | - Julin Xiao
- Research Center for Novel Computing Sensing and Intelligent Processing, Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
| | - Yao Yang
- Research Center for Space Computing System, Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
| | - Wei Guo
- School of Materials Science and Engineering, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212100, China
| | - Yueming Wang
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
- Key Laboratory of Space Active Optoelectronics Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, China
| | - Jing Jiang
- Research Center for High Efficiency Computing System, Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
25
|
Yeshi K, Jamtsho T, Wangchuk P. Current Treatments, Emerging Therapeutics, and Natural Remedies for Inflammatory Bowel Disease. Molecules 2024; 29:3954. [PMID: 39203033 PMCID: PMC11357616 DOI: 10.3390/molecules29163954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, lifelong disorder characterized by inflammation of the gastrointestinal (GI) tract. The exact etiology of IBD remains incompletely understood due to its multifaceted nature, which includes genetic predisposition, environmental factors, and host immune response dysfunction. Currently, there is no cure for IBD. This review discusses the available treatment options and the challenges they present. Importantly, we examine emerging therapeutics, such as biologics and immunomodulators, that offer targeted treatment strategies for IBD. While many IBD patients do not respond adequately to most biologics, recent clinical trials combining biologics with small-molecule drugs (SMDs) have provided new insights into improving the IBD treatment landscape. Furthermore, numerous novel and specific therapeutic targets have been identified. The high cost of IBD drugs poses a significant barrier to treatment, but this challenge may be alleviated with the development of more affordable biosimilars. Additionally, emerging point-of-care protein biomarkers from serum and plasma are showing potential for enhancing the precision of IBD diagnosis and prognosis. Several natural products (NPs), including crude extracts, small molecules, and peptides, have demonstrated promising anti-inflammatory activity in high-throughput screening (HTS) systems and advanced artificial intelligence (AI)-assisted platforms, such as molecular docking and ADMET prediction. These platforms are advancing the search for alternative IBD therapies derived from natural sources, potentially leading to more affordable and safer treatment options with fewer side effects.
Collapse
Affiliation(s)
- Karma Yeshi
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia;
- Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia
| | - Tenzin Jamtsho
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia;
- Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia
| | - Phurpa Wangchuk
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia;
- Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia
| |
Collapse
|
26
|
Zhang WY, Zheng XL, Coghi PS, Chen JH, Dong BJ, Fan XX. Revolutionizing adjuvant development: harnessing AI for next-generation cancer vaccines. Front Immunol 2024; 15:1438030. [PMID: 39206192 PMCID: PMC11349682 DOI: 10.3389/fimmu.2024.1438030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
With the COVID-19 pandemic, the importance of vaccines has been widely recognized and has led to increased research and development efforts. Vaccines also play a crucial role in cancer treatment by activating the immune system to target and destroy cancer cells. However, enhancing the efficacy of cancer vaccines remains a challenge. Adjuvants, which enhance the immune response to antigens and improve vaccine effectiveness, have faced limitations in recent years, resulting in few novel adjuvants being identified. The advancement of artificial intelligence (AI) technology in drug development has provided a foundation for adjuvant screening and application, leading to a diversification of adjuvants. This article reviews the significant role of tumor vaccines in basic research and clinical treatment and explores the use of AI technology to screen novel adjuvants from databases. The findings of this review offer valuable insights for the development of new adjuvants for next-generation vaccines.
Collapse
Affiliation(s)
- Wan-Ying Zhang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Xiao-Li Zheng
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Paolo Saul Coghi
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jun-Hui Chen
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bing-Jun Dong
- Gynecology Department, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, China
| | - Xing-Xing Fan
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| |
Collapse
|
27
|
Menke J, Nahal Y, Bjerrum EJ, Kabeshov M, Kaski S, Engkvist O. Metis: a python-based user interface to collect expert feedback for generative chemistry models. J Cheminform 2024; 16:100. [PMID: 39143631 PMCID: PMC11323385 DOI: 10.1186/s13321-024-00892-3] [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: 05/15/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024] Open
Abstract
One challenge that current de novo drug design models face is a disparity between the user's expectations and the actual output of the model in practical applications. Tailoring models to better align with chemists' implicit knowledge, expectation and preferences is key to overcoming this obstacle effectively. While interest in preference-based and human-in-the-loop machine learning in chemistry is continuously increasing, no tool currently exists that enables the collection of standardized and chemistry-specific feedback. Metis is a Python-based open-source graphical user interface (GUI), designed to solve this and enable the collection of chemists' detailed feedback on molecular structures. The GUI enables chemists to explore and evaluate molecules, offering a user-friendly interface for annotating preferences and specifying desired or undesired structural features. By providing chemists the opportunity to give detailed feedback, allows researchers to capture more efficiently the chemist's implicit knowledge and preferences. This knowledge is crucial to align the chemist's idea with the de novo design agents. The GUI aims to enhance this collaboration between the human and the "machine" by providing an intuitive platform where chemists can interactively provide feedback on molecular structures, aiding in preference learning and refining de novo design strategies. Metis integrates with the existing de novo framework REINVENT, creating a closed-loop system where human expertise can continuously inform and refine the generative models.Scientific contributionWe introduce a novel Graphical User Interface, that allows chemists/researchers to give detailed feedback on substructures and properties of small molecules. This tool can be used to learn the preferences of chemists in order to align de novo drug design models with the chemist's ideas. The GUI can be customized to fit different needs and projects and enables direct integration into de novo REINVENT runs. We believe that Metis can facilitate the discussion and development of novel ways to integrate human feedback that goes beyond binary decisions of liking or disliking a molecule.
Collapse
Affiliation(s)
- Janosch Menke
- Department of Computer Science and Engineering, Chalmers University of Technology, Gothenburg, 41296, Sweden.
| | - Yasmine Nahal
- Department of Computer Science, Aalto University, Espoo, 02150, Finland
| | | | - Mikhail Kabeshov
- Molecular AI, Discovery Sciences AstraZeneca R &D, Mölndal, 43183, Sweden
| | - Samuel Kaski
- Department of Computer Science, Aalto University, Espoo, 02150, Finland
- Department of Computer Science, University of Manchester, Manchester, M13 9PL, UK
| | - Ola Engkvist
- Department of Computer Science and Engineering, Chalmers University of Technology, Gothenburg, 41296, Sweden
- Molecular AI, Discovery Sciences AstraZeneca R &D, Mölndal, 43183, Sweden
| |
Collapse
|
28
|
Odena C, Santiago TG, Linares ML, Castellanos-Blanco N, McGuire RT, Chaves-Arquero B, Alonso JM, Diéguez-Vázquez A, Tan E, Alcázar J, Buijnsters P, Cañellas S, Martin R. Late-Stage C( sp2)-C( sp3) Diversification via Nickel Oxidative Addition Complexes. J Am Chem Soc 2024; 146:21264-21270. [PMID: 39052124 DOI: 10.1021/jacs.4c08404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Herein, we describe nickel oxidative addition complexes (Ni-OACs) of drug-like molecules as a platform to rapidly generate lead candidates with enhanced C(sp3) fraction. The potential of Ni-OACs to access new chemical space has been assessed not only in C(sp2)-C(sp3) couplings but also in additional bond formations without recourse to specialized ligands and with improved generality when compared to Ni-catalyzed reactions. The development of an automated diversification process further illustrates the robustness of Ni-OACs, thus offering a new gateway to expedite the design-make-test-analyze (DMTA) cycle in drug discovery.
Collapse
Affiliation(s)
- Carlota Odena
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, Avenida Països Catalans 16, 43007 Tarragona, Spain
- Departament de Química Orgànica, Universitat Rovira i Virgili, c/Marcel·lí Domingo 1, 43007 Tarragona, Spain
| | - Tomás G Santiago
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, Avenida Països Catalans 16, 43007 Tarragona, Spain
| | | | - Nahury Castellanos-Blanco
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, Avenida Països Catalans 16, 43007 Tarragona, Spain
| | - Ryan T McGuire
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, Avenida Països Catalans 16, 43007 Tarragona, Spain
| | - Belén Chaves-Arquero
- Janssen-Cilag, S.A., a Johnson & Johnson Company, C/Jarama 75A, 45007 Toledo, Spain
| | - Jose Manuel Alonso
- Janssen-Cilag, S.A., a Johnson & Johnson Company, C/Jarama 75A, 45007 Toledo, Spain
| | | | - Eric Tan
- Janssen Pharmaceutica Nv, A Johnson & Johnson Company, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Jesús Alcázar
- Janssen-Cilag, S.A., a Johnson & Johnson Company, C/Jarama 75A, 45007 Toledo, Spain
| | - Peter Buijnsters
- Janssen Pharmaceutica Nv, A Johnson & Johnson Company, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Santiago Cañellas
- Janssen-Cilag, S.A., a Johnson & Johnson Company, C/Jarama 75A, 45007 Toledo, Spain
| | - Ruben Martin
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, Avenida Països Catalans 16, 43007 Tarragona, Spain
- ICREA, Passeig Lluís Companys, 23, 08010 Barcelona, Spain
| |
Collapse
|
29
|
Sagmeister P, Melnizky L, Williams JD, Kappe CO. Simultaneous reaction- and analytical model building using dynamic flow experiments to accelerate process development. Chem Sci 2024; 15:12523-12533. [PMID: 39118626 PMCID: PMC11304546 DOI: 10.1039/d4sc01703j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/29/2024] [Indexed: 08/10/2024] Open
Abstract
In modern pharmaceutical research, the demand for expeditious development of synthetic routes to active pharmaceutical ingredients (APIs) has led to a paradigm shift towards data-rich process development. Conventional methodologies encompass prolonged timelines for the development of both a reaction model and analytical models. The development of both methods are often separated into different departments and can require an iterative optimization process. Addressing this issue, we introduce an innovative dual modeling approach, combining the development of a Process Analytical Technology (PAT) strategy with reaction optimization. This integrated approach is exemplified in diverse amidation reactions and the synthesis of the API benznidazole. The platform, characterized by a high degree of automation and minimal operator involvement, achieves PAT calibration through a "standard addition" approach. Dynamic experiments are executed to screen a broad process space and gather data for fitting kinetic parameters. Employing an open-source software program facilitates rapid kinetic parameter fitting and additional in silico optimization within minutes. This highly automated workflow not only expedites the understanding and optimization of chemical processes, but also holds significant promise for time and resource savings within the pharmaceutical industry.
Collapse
Affiliation(s)
- Peter Sagmeister
- Institute of Chemistry, University of Graz, NAWI Graz Heinrichstrasse 28 8010 Graz Austria
- Center for Continuous Flow Synthesis and Processing (CC FLOW), Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13 8010 Graz Austria
| | - Lukas Melnizky
- Institute of Chemistry, University of Graz, NAWI Graz Heinrichstrasse 28 8010 Graz Austria
- Center for Continuous Flow Synthesis and Processing (CC FLOW), Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13 8010 Graz Austria
| | - Jason D Williams
- Institute of Chemistry, University of Graz, NAWI Graz Heinrichstrasse 28 8010 Graz Austria
- Center for Continuous Flow Synthesis and Processing (CC FLOW), Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13 8010 Graz Austria
| | - C Oliver Kappe
- Institute of Chemistry, University of Graz, NAWI Graz Heinrichstrasse 28 8010 Graz Austria
- Center for Continuous Flow Synthesis and Processing (CC FLOW), Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13 8010 Graz Austria
| |
Collapse
|
30
|
Gholap AD, Uddin MJ, Faiyazuddin M, Omri A, Gowri S, Khalid M. Advances in artificial intelligence for drug delivery and development: A comprehensive review. Comput Biol Med 2024; 178:108702. [PMID: 38878397 DOI: 10.1016/j.compbiomed.2024.108702] [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: 01/03/2024] [Revised: 05/12/2024] [Accepted: 06/01/2024] [Indexed: 07/24/2024]
Abstract
Artificial intelligence (AI) has emerged as a powerful tool to revolutionize the healthcare sector, including drug delivery and development. This review explores the current and future applications of AI in the pharmaceutical industry, focusing on drug delivery and development. It covers various aspects such as smart drug delivery networks, sensors, drug repurposing, statistical modeling, and simulation of biotechnological and biological systems. The integration of AI with nanotechnologies and nanomedicines is also examined. AI offers significant advancements in drug discovery by efficiently identifying compounds, validating drug targets, streamlining drug structures, and prioritizing response templates. Techniques like data mining, multitask learning, and high-throughput screening contribute to better drug discovery and development innovations. The review discusses AI applications in drug formulation and delivery, clinical trials, drug safety, and pharmacovigilance. It addresses regulatory considerations and challenges associated with AI in pharmaceuticals, including privacy, data security, and interpretability of AI models. The review concludes with future perspectives, highlighting emerging trends, addressing limitations and biases in AI models, and emphasizing the importance of collaboration and knowledge sharing. It provides a comprehensive overview of AI's potential to transform the pharmaceutical industry and improve patient care while identifying further research and development areas.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar, Maharashtra, 401404, India.
| | - Md Jasim Uddin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Md Faiyazuddin
- School of Pharmacy, Al-Karim University, Katihar, Bihar, 854106, India; Centre for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Tamil Nadu, India.
| | - Abdelwahab Omri
- Department of Chemistry and Biochemistry, The Novel Drug and Vaccine Delivery Systems Facility, Laurentian University, Sudbury, ON, P3E 2C6, Canada.
| | - S Gowri
- PG & Research, Department of Physics, Cauvery College for Women, Tiruchirapalli, Tamil Nadu, 620018, India
| | - Mohammad Khalid
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; Sunway Centre for Electrochemical Energy and Sustainable Technology (SCEEST), School of Engineering and Technology, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia; University Centre for Research and Development, Chandigarh University, Mohali, Punjab, 140413, India.
| |
Collapse
|
31
|
Zhang R, Zhang C, Fan X, Au Yeung CCK, Li H, Lin H, Shum HC. A droplet robotic system enabled by electret-induced polarization on droplet. Nat Commun 2024; 15:6220. [PMID: 39043732 PMCID: PMC11266649 DOI: 10.1038/s41467-024-50520-9] [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: 12/04/2023] [Accepted: 07/12/2024] [Indexed: 07/25/2024] Open
Abstract
Robotics for scientific research are evolving from grasping macro-scale solid materials to directly actuating micro-scale liquid samples. However, current liquid actuation mechanisms often restrict operable liquid types or compromise the activity of biochemical samples by introducing interfering mediums. Here, we propose a robotic liquid handling system enabled by a novel droplet actuation mechanism, termed electret-induced polarization on droplet (EPD). EPD enables all-liquid actuation in principle and experimentally exhibits generality for actuating various inorganic/organic liquids with relative permittivity ranging from 2.25 to 84.2 and volume from 500 nL to 1 mL. Moreover, EPD is capable of actuating various biochemical samples without compromising their activities, including various body fluids, living cells, and proteins. A robotic system is also coupled with the EPD mechanism to enable full automation. EPD's high adaptability with liquid types and biochemical samples thus promotes the automation of liquid-based scientific experiments across multiple disciplines.
Collapse
Affiliation(s)
- Ruotong Zhang
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong SAR, China
| | - Chengzhi Zhang
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong SAR, China
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xiaoxue Fan
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong SAR, China
| | - Christina C K Au Yeung
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong SAR, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Hong Kong SAR, China
| | - Huiyanchen Li
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Hong Kong SAR, China
| | - Haisong Lin
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong SAR, China.
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Hong Kong SAR, China.
| | - Ho Cheung Shum
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong SAR, China.
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Hong Kong SAR, China.
| |
Collapse
|
32
|
Mervin L, Voronov A, Kabeshov M, Engkvist O. QSARtuna: An Automated QSAR Modeling Platform for Molecular Property Prediction in Drug Design. J Chem Inf Model 2024; 64:5365-5374. [PMID: 38950185 DOI: 10.1021/acs.jcim.4c00457] [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: 07/03/2024]
Abstract
Machine-learning (ML) and deep-learning (DL) approaches to predict the molecular properties of small molecules are increasingly deployed within the design-make-test-analyze (DMTA) drug design cycle to predict molecular properties of interest. Despite this uptake, there are only a few automated packages to aid their development and deployment that also support uncertainty estimation, model explainability, and other key aspects of model usage. This represents a key unmet need within the field, and the large number of molecular representations and algorithms (and associated parameters) means it is nontrivial to robustly optimize, evaluate, reproduce, and deploy models. Here, we present QSARtuna, a molecule property prediction modeling pipeline, written in Python and utilizing the Optuna, Scikit-learn, RDKit, and ChemProp packages, which enables the efficient and automated comparison between molecular representations and machine learning models. The platform was developed by considering the increasingly important aspect of model uncertainty quantification and explainability by design. We provide details for our framework and provide illustrative examples to demonstrate the capability of the software when applied to simple molecular property, reaction/reactivity prediction, and DNA encoded library enrichment classification. We hope that the release of QSARtuna will further spur innovation in automatic ML modeling and provide a platform for education of best practices in molecular property modeling. The code for the QSARtuna framework is made freely available via GitHub.
Collapse
Affiliation(s)
- Lewis Mervin
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, United Kingdom
| | - Alexey Voronov
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg 412 96, Sweden
| | - Mikhail Kabeshov
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg 412 96, Sweden
| | - Ola Engkvist
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg 412 96, Sweden
- Department of Computer Science and Engineering, University of Gothenburg, Chalmers University of Technology, Gothenburg 412 96, Sweden
| |
Collapse
|
33
|
Dong Y, Wang J, Chen L, Chen H, Dang S, Li F. Aptamer-based assembly systems for SARS-CoV-2 detection and therapeutics. Chem Soc Rev 2024; 53:6830-6859. [PMID: 38829187 DOI: 10.1039/d3cs00774j] [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: 06/05/2024]
Abstract
Nucleic acid aptamers are oligonucleotide chains with molecular recognition properties. Compared with antibodies, aptamers show advantages given that they are readily produced via chemical synthesis and elicit minimal immunogenicity in biomedicine applications. Notably, aptamer-encoded nucleic acid assemblies further improve the binding affinity of aptamers with the targets due to their multivalent synergistic interactions. Specially, aptamers can be engineered with special topological arrangements in nucleic acid assemblies, which demonstrate spatial and valence matching towards antigens on viruses, thus showing potential in the detection and therapeutic applications of viruses. This review presents the recent progress on the aptamers explored for SARS-CoV-2 detection and infection treatment, wherein applications of aptamer-based assembly systems are introduced in detail. Screening methods and chemical modification strategies for aptamers are comprehensively summarized, and the types of aptamers employed against different target domains of SARS-CoV-2 are illustrated. The evolution of aptamer-based assembly systems for the detection and neutralization of SARS-CoV-2, as well as the construction principle and characteristics of aptamer-based DNA assemblies are demonstrated. The typically representative works are presented to demonstrate how to assemble aptamers rationally and elaborately for specific applications in SARS-CoV-2 diagnosis and neutralization. Finally, we provide deep insights into the current challenges and future perspectives towards aptamer-based nucleic acid assemblies for virus detection and neutralization in nanomedicine.
Collapse
Affiliation(s)
- Yuhang Dong
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.
| | - Jingping Wang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.
| | - Ling Chen
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.
| | - Haonan Chen
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.
| | - Shuangbo Dang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.
| | - Feng Li
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.
| |
Collapse
|
34
|
Calado CRC. Bridging the gap between target-based and phenotypic-based drug discovery. Expert Opin Drug Discov 2024; 19:789-798. [PMID: 38747562 DOI: 10.1080/17460441.2024.2355330] [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: 09/05/2023] [Accepted: 05/10/2024] [Indexed: 06/26/2024]
Abstract
INTRODUCTION The unparalleled progress in science of the last decades has brought a better understanding of the molecular mechanisms of diseases. This promoted drug discovery processes based on a target approach. However, despite the high promises associated, a critical decrease in the number of first-in-class drugs has been observed. AREAS COVERED This review analyses the challenges, advances, and opportunities associated with the main strategies of the drug discovery process, i.e. based on a rational target approach and on an empirical phenotypic approach. This review also evaluates how the gap between these two crossroads can be bridged toward a more efficient drug discovery process. EXPERT OPINION The critical lack of knowledge of the complex biological networks is leading to targets not relevant for the clinical context or to drugs that present undesired adverse effects. The phenotypic systems designed by considering available molecular mechanisms can mitigate these knowledge gaps. Associated with the expansion of the chemical space and other technologies, these designs can lead to more efficient drug discoveries. Technological and scientific knowledge should also be applied to identify, as early as possible, both drug targets and mechanisms of action, leading to a more efficient drug discovery pipeline.
Collapse
Affiliation(s)
- Cecília R C Calado
- ISEL-Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Lisboa, Portugal
- iBB - Institute for Bioengineering and Biosciences, i4HB - The Associate Laboratory Institute for Health and Bioeconomy, IST - Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
35
|
Wu NP, Wang W, Gadiagellan D, Counsell M, Hamidi NK, Koike Y, Nguyen HQ. An Automated Robotic Interface for Assays: Facilitating Machine Learning in Drug Discovery by the Automation of Physicochemical Property Assays. ACS OMEGA 2024; 9:24948-24958. [PMID: 38882107 PMCID: PMC11170699 DOI: 10.1021/acsomega.4c02003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024]
Abstract
Measuring the physicochemical properties of molecules is an iterative but integral process in the drug development process. A strategy to overcome the challenges in maximizing assay throughput relies on the usage of in silico machine learning (ML) prediction models trained on experimental data. Consequently, the performance of these in silico models are dependent on the quality of the utilized experimental data. To improve the data quality, we have designed and implemented an automated robotic system to prepare and run physicochemical property assays (Automated Robotic Interface for Assays, ARIA) with an increase in sample throughput of 6 to10-fold. Through this process, we overcame major challenges and achieved consistent reproducible assay data compared to semiautomated assay preparation.
Collapse
Affiliation(s)
- Newton P Wu
- Analytical Research, Discovery Chemistry Department, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Wenyi Wang
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | | | - Mike Counsell
- UK Robotics, Inc., Manchester BL5 3EH, United Kingdom
| | - Nikkia K Hamidi
- Analytical Research, Discovery Chemistry Department, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Yuko Koike
- Analytical Research, Discovery Chemistry Department, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Huy Q Nguyen
- Analytical Research, Discovery Chemistry Department, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
36
|
Bassani D, Parrott NJ, Manevski N, Zhang JD. Another string to your bow: machine learning prediction of the pharmacokinetic properties of small molecules. Expert Opin Drug Discov 2024; 19:683-698. [PMID: 38727016 DOI: 10.1080/17460441.2024.2348157] [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: 10/23/2023] [Accepted: 04/23/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Prediction of pharmacokinetic (PK) properties is crucial for drug discovery and development. Machine-learning (ML) models, which use statistical pattern recognition to learn correlations between input features (such as chemical structures) and target variables (such as PK parameters), are being increasingly used for this purpose. To embed ML models for PK prediction into workflows and to guide future development, a solid understanding of their applicability, advantages, limitations, and synergies with other approaches is necessary. AREAS COVERED This narrative review discusses the design and application of ML models to predict PK parameters of small molecules, especially in light of established approaches including in vitro-in vivo extrapolation (IVIVE) and physiologically based pharmacokinetic (PBPK) models. The authors illustrate scenarios in which the three approaches are used and emphasize how they enhance and complement each other. In particular, they highlight achievements, the state of the art and potentials of applying machine learning for PK prediction through a comphrehensive literature review. EXPERT OPINION ML models, when carefully crafted, regularly updated, and appropriately used, empower users to prioritize molecules with favorable PK properties. Informed practitioners can leverage these models to improve the efficiency of drug discovery and development process.
Collapse
Affiliation(s)
- Davide Bassani
- Pharmaceutical Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Neil John Parrott
- Pharmaceutical Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Nenad Manevski
- Pharmaceutical Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Jitao David Zhang
- Pharmaceutical Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
37
|
Qin T, Hernandez SER, Shiers J, Crittall M, Novak A, Smith CS. A decentralized solid compound storage facility managed by a centralized electronic platform at a growing drug discovery company. SLAS Technol 2024; 29:100143. [PMID: 38740284 DOI: 10.1016/j.slast.2024.100143] [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: 11/18/2023] [Revised: 03/13/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
Within a growing drug discovery company, scientists acquire (either through in house synthesis or purchase) then store, retrieve, and ship solid compound samples daily between multiple locations. The efficient management and tracking of this entire process to support drug discovery is a significant challenge. This article describes a decentralized and cost-effective inventory facility that simplifies the solid compound storage and retrieval process. Standardized storage cabinets from the market are utilized, providing a cost-effective physical infrastructure. The cabinets can be distributed across storage rooms at multiple sites and arranged into spaces with a variety of dimensions, allowing the system to be retrofitted into existing facilities and scaled up easily. We can provide storage close to work areas at each location, minimizing both unnecessary movement of staff and transportation of substances. We have applied a systematic barcoding method to the compound batch identifier that correlates with its compound location. This simplifies the compound registration process as well as the process of finding and returning compounds. Additionally, a centralized electronic platform has been employed to store, update and track solid compound information, such as properties, location and quantity. Compound shipment may be initiated from different sites, and a centralized electronic platform assists the information retrieval process, ensuring each location possesses up-to-date information. The electronic platform we present streamlines the management of compound registration, location tracking, weight updates and shipment information, facilitating seamless record sharing among all stakeholders. Every step of the process can be tracked in real time by the project team. The platform can be flexibly configured to adapt to an evolving set of storage locations, with all information and processes being audited.
Collapse
Affiliation(s)
- Ting Qin
- Sygnature Discovery Ltd., BioCity, Pennyfoot Street, Nottingham, United Kingdom.
| | | | - Jason Shiers
- Sygnature Discovery Ltd., BioCity, Pennyfoot Street, Nottingham, United Kingdom
| | - Matthew Crittall
- Sygnature Discovery Ltd., BioCity, Pennyfoot Street, Nottingham, United Kingdom
| | - Andrew Novak
- Sygnature Discovery Ltd., BioCity, Pennyfoot Street, Nottingham, United Kingdom
| | | |
Collapse
|
38
|
Wang L, Zhou Z, Yang X, Shi S, Zeng X, Cao D. The present state and challenges of active learning in drug discovery. Drug Discov Today 2024; 29:103985. [PMID: 38642700 DOI: 10.1016/j.drudis.2024.103985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
Active learning (AL) is an iterative feedback process that efficiently identifies valuable data within vast chemical space, even with limited labeled data. This characteristic renders it a valuable approach to tackle the ongoing challenges faced in drug discovery, such as the ever-expanding explore space and the limitations of labeled data. Consequently, AL is increasingly gaining prominence in the field of drug development. In this paper, we comprehensively review the application of AL at all stages of drug discovery, including compounds-target interaction prediction, virtual screening, molecular generation and optimization, as well as molecular properties prediction. Additionally, we discuss the challenges and prospects associated with the current applications of AL in drug discovery.
Collapse
Affiliation(s)
- Lei Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Zhenran Zhou
- Department of Computer Science, Hunan University, Changsha 410082, Hunan, China
| | - Xixi Yang
- Department of Computer Science, Hunan University, Changsha 410082, Hunan, China
| | - Shaohua Shi
- Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiangxiang Zeng
- Department of Computer Science, Hunan University, Changsha 410082, Hunan, China.
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
39
|
Kamya P, Ozerov IV, Pun FW, Tretina K, Fokina T, Chen S, Naumov V, Long X, Lin S, Korzinkin M, Polykovskiy D, Aliper A, Ren F, Zhavoronkov A. PandaOmics: An AI-Driven Platform for Therapeutic Target and Biomarker Discovery. J Chem Inf Model 2024; 64:3961-3969. [PMID: 38404138 PMCID: PMC11134400 DOI: 10.1021/acs.jcim.3c01619] [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: 10/10/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/27/2024]
Abstract
PandaOmics is a cloud-based software platform that applies artificial intelligence and bioinformatics techniques to multimodal omics and biomedical text data for therapeutic target and biomarker discovery. PandaOmics generates novel and repurposed therapeutic target and biomarker hypotheses with the desired properties and is available through licensing or collaboration. Targets and biomarkers generated by the platform were previously validated in both in vitro and in vivo studies. PandaOmics is a core component of Insilico Medicine's Pharma.ai drug discovery suite, which also includes Chemistry42 for the de novo generation of novel small molecules, and inClinico─a data-driven multimodal platform that forecasts a clinical trial's probability of successful transition from phase 2 to phase 3. In this paper, we demonstrate how the PandaOmics platform can efficiently identify novel molecular targets and biomarkers for various diseases.
Collapse
Affiliation(s)
- Petrina Kamya
- Insilico
Medicine Canada Inc., 3710-1250 René-Lévesque Blvd. W, Montreal, Quebec, Canada H3B 4W8
| | - Ivan V. Ozerov
- Insilico
Medicine Hong Kong Limited, Hong Kong Science and Technology Park, Hong Kong
| | - Frank W. Pun
- Insilico
Medicine Hong Kong Limited, Hong Kong Science and Technology Park, Hong Kong
| | - Kyle Tretina
- Insilico
Medicine Hong Kong Limited, Hong Kong Science and Technology Park, Hong Kong
| | - Tatyana Fokina
- Insilico
Medicine Hong Kong Limited, Hong Kong Science and Technology Park, Hong Kong
| | - Shan Chen
- Insilico
Medicine Shanghai Limited, Suite 901, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai 201203, China
| | - Vladimir Naumov
- Insilico
Medicine Hong Kong Limited, Hong Kong Science and Technology Park, Hong Kong
| | - Xi Long
- Insilico
Medicine Hong Kong Limited, Hong Kong Science and Technology Park, Hong Kong
| | - Sha Lin
- Insilico
Medicine Shanghai Limited, Suite 901, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai 201203, China
| | - Mikhail Korzinkin
- Insilico
Medicine Hong Kong Limited, Hong Kong Science and Technology Park, Hong Kong
| | - Daniil Polykovskiy
- Insilico
Medicine Canada Inc., 3710-1250 René-Lévesque Blvd. W, Montreal, Quebec, Canada H3B 4W8
| | - Alex Aliper
- Insilico
Medicine AI Limited, Level 6, Unit 08, Block A, IRENA HQ Building, P.O.
Box 145748, Masdar City, Abu Dhabi, United Arab Emirates
| | - Feng Ren
- Insilico
Medicine Shanghai Limited, Suite 901, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai 201203, China
| | - Alex Zhavoronkov
- Insilico
Medicine Hong Kong Limited, Hong Kong Science and Technology Park, Hong Kong
- Insilico
Medicine AI Limited, Level 6, Unit 08, Block A, IRENA HQ Building, P.O.
Box 145748, Masdar City, Abu Dhabi, United Arab Emirates
- Buck
Institute for Research on Aging, Novato, California 94945, United States
| |
Collapse
|
40
|
Rovenchak A, Druchok M. Machine learning-assisted search for novel coagulants: When machine learning can be efficient even if data availability is low. J Comput Chem 2024; 45:937-952. [PMID: 38174834 DOI: 10.1002/jcc.27292] [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: 10/30/2023] [Revised: 12/04/2023] [Accepted: 12/10/2023] [Indexed: 01/05/2024]
Abstract
Design of new drugs is a challenging process: a candidate molecule should satisfy multiple conditions to act properly and make the least side-effect-perfect candidates selectively attach to and influence only targets, leaving off-targets intact. The amount of experimental data about various properties of molecules constantly grows, promoting data-driven approaches. However, the applicability of typical predictive machine learning techniques can be substantially limited by a lack of experimental data about a particular target. For example, there are many known Thrombin inhibitors (acting as anticoagulants), but a very limited number of known Protein C inhibitors (coagulants). In this study, we present our approach to suggest new inhibitor candidates by building an effective representation of chemical space. For this aim, we developed a deep learning model-autoencoder, trained on a large set of molecules in the SMILES format to map the chemical space. Further, we applied different sampling strategies to generate novel coagulant candidates. Symmetrically, we tested our approach on anticoagulant candidates, where we were able to predict their inhibition towards Thrombin. We also compare our approach with MegaMolBART-another deep learning generative model, but exploiting similar principles of navigation in a chemical space.
Collapse
Affiliation(s)
- Andrij Rovenchak
- SoftServe, Inc., Lviv, Ukraine
- Professor Ivan Vakarchuk Department for Theoretical Physics, Ivan Franko National University of Lviv, Lviv, Ukraine
| | - Maksym Druchok
- SoftServe, Inc., Lviv, Ukraine
- Institute for Condensed Matter Physics, Lviv, Ukraine
| |
Collapse
|
41
|
Oniani D, Hilsman J, Zang C, Wang J, Cai L, Zawala J, Wang Y. Emerging opportunities of using large language models for translation between drug molecules and indications. Sci Rep 2024; 14:10738. [PMID: 38730226 PMCID: PMC11087469 DOI: 10.1038/s41598-024-61124-0] [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: 02/14/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024] Open
Abstract
A drug molecule is a substance that changes an organism's mental or physical state. Every approved drug has an indication, which refers to the therapeutic use of that drug for treating a particular medical condition. While the Large Language Model (LLM), a generative Artificial Intelligence (AI) technique, has recently demonstrated effectiveness in translating between molecules and their textual descriptions, there remains a gap in research regarding their application in facilitating the translation between drug molecules and indications (which describes the disease, condition or symptoms for which the drug is used), or vice versa. Addressing this challenge could greatly benefit the drug discovery process. The capability of generating a drug from a given indication would allow for the discovery of drugs targeting specific diseases or targets and ultimately provide patients with better treatments. In this paper, we first propose a new task, the translation between drug molecules and corresponding indications, and then test existing LLMs on this new task. Specifically, we consider nine variations of the T5 LLM and evaluate them on two public datasets obtained from ChEMBL and DrugBank. Our experiments show the early results of using LLMs for this task and provide a perspective on the state-of-the-art. We also emphasize the current limitations and discuss future work that has the potential to improve the performance on this task. The creation of molecules from indications, or vice versa, will allow for more efficient targeting of diseases and significantly reduce the cost of drug discovery, with the potential to revolutionize the field of drug discovery in the era of generative AI.
Collapse
Affiliation(s)
- David Oniani
- Department of Health Information Management, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jordan Hilsman
- Department of Health Information Management, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chengxi Zang
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, New York, NY, USA
| | - Junmei Wang
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lianjin Cai
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jan Zawala
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Kraków, Poland
| | - Yanshan Wang
- Department of Health Information Management, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA.
- Intelligent Systems Program, University of Pittsburgh, Pittsburgh, PA, USA.
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
42
|
Kench T, Rahardjo A, Terrones GG, Bellamkonda A, Maher TE, Storch M, Kulik HJ, Vilar R. A Semi-Automated, High-Throughput Approach for the Synthesis and Identification of Highly Photo-Cytotoxic Iridium Complexes. Angew Chem Int Ed Engl 2024; 63:e202401808. [PMID: 38404222 DOI: 10.1002/anie.202401808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
The discovery of new compounds with pharmacological properties is usually a lengthy, laborious and expensive process. Thus, there is increasing interest in developing workflows that allow for the rapid synthesis and evaluation of libraries of compounds with the aim of identifying leads for further drug development. Herein, we apply combinatorial synthesis to build a library of 90 iridium(III) complexes (81 of which are new) over two synthesise-and-test cycles, with the aim of identifying potential agents for photodynamic therapy. We demonstrate the power of this approach by identifying highly active complexes that are well-tolerated in the dark but display very low nM phototoxicity against cancer cells. To build a detailed structure-activity relationship for this class of compounds we have used density functional theory (DFT) calculations to determine some key electronic parameters and study correlations with the experimental data. Finally, we present an optimised semi-automated synthesise-and-test protocol to obtain multiplex data within 72 hours.
Collapse
Affiliation(s)
- Timothy Kench
- Department of Chemistry, Imperial College London, White City Campus, W12 0BZ, London, UK
| | - Arielle Rahardjo
- Department of Chemistry, Imperial College London, White City Campus, W12 0BZ, London, UK
| | - Gianmarco G Terrones
- Department of Chemical Engineering, Massachusetts Institute of Technology, 02139, Cambridge, MA, USA
| | | | - Thomas E Maher
- Department of Chemistry, Imperial College London, White City Campus, W12 0BZ, London, UK
- Institute of Chemical Biology, Imperial College London, White City Campus, W12 0BZ, London, UK
| | - Marko Storch
- Department of Infectious Disease, Imperial College London, South Kensington Campus, SW7 2AZ, London, UK
- London Biofoundry, Imperial College Translation and Innovation Hub, W12 0BZ, London, UK
| | - Heather J Kulik
- Department of Chemical Engineering, Massachusetts Institute of Technology, 02139, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, 02139, Cambridge, MA, USA
| | - Ramon Vilar
- Department of Chemistry, Imperial College London, White City Campus, W12 0BZ, London, UK
- Institute of Chemical Biology, Imperial College London, White City Campus, W12 0BZ, London, UK
| |
Collapse
|
43
|
Mohammadi M, Ahmed Qadir S, Mahmood Faraj A, Hamid Shareef O, Mahmoodi H, Mahmoudi F, Moradi S. Navigating the future: Microfluidics charting new routes in drug delivery. Int J Pharm 2024:124142. [PMID: 38648941 DOI: 10.1016/j.ijpharm.2024.124142] [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: 10/12/2023] [Revised: 03/30/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Microfluidics has emerged as a transformative force in the field of drug delivery, offering innovative avenues to produce a diverse range of nano drug delivery systems. Thanks to its precise manipulation of small fluid volumes and its exceptional command over the physicochemical characteristics of nanoparticles, this technology is notably able to enhance the pharmacokinetics of drugs. It has initiated a revolutionary phase in the domain of drug delivery, presenting a multitude of compelling advantages when it comes to developing nanocarriers tailored for the delivery of poorly soluble medications. These advantages represent a substantial departure from conventional drug delivery methodologies, marking a paradigm shift in pharmaceutical research and development. Furthermore, microfluidic platformsmay be strategically devised to facilitate targeted drug delivery with the objective of enhancing the localized bioavailability of pharmaceutical substances. In this paper, we have comprehensively investigated a range of significant microfluidic techniques used in the production of nanoscale drug delivery systems. This comprehensive review can serve as a valuable reference and offer insightful guidance for the development and optimization of numerous microfluidics-fabricated nanocarriers.
Collapse
Affiliation(s)
- Mohammad Mohammadi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Syamand Ahmed Qadir
- Department of Medical Laboratory Techniques, Halabja Technical Institute, Research Center, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
| | - Aryan Mahmood Faraj
- Department of Medical Laboratory Sciences, Halabja Technical College of Applied Sciences, Sulaimani Polytechnic University, Halabja, Iraq
| | - Osama Hamid Shareef
- Department of Medical Laboratory Techniques, Halabja Technical Institute, Research Center, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
| | - Hassan Mahmoodi
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mahmoudi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sajad Moradi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
44
|
Faizan S, Talath S, Wali AF, Hani U, Haider N, Mandal SP, Kumar BRP. Anticancer potential of novel symmetrical and asymmetrical dihydropyridines against breast cancer via EGFR inhibition: molecular design, synthesis, analysis and screening. RSC Adv 2024; 14:11368-11387. [PMID: 38595721 PMCID: PMC11002980 DOI: 10.1039/d4ra01424c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 03/29/2024] [Indexed: 04/11/2024] Open
Abstract
A series of novel symmetrical and asymmetrical dihydropyridines (HD 1-15) were designed, subjected to in silico ADMET prediction, synthesized, analyzed by IR, NMR, Mass analytical techniques and evaluated against epidermal growth factor receptor (EGFR) as inhibitors against Breast cancer. The results of predicted ADMET studies demonstrated the drug-likeness properties of the reported compounds. The in vitro cytotoxicity assessment of the synthesized compounds revealed that all of them showed good activity (IC50 ranging from 16.75 to 66.54 μM) towards MCF-7 breast cancer cells compared to the standard drug, Lapatinib (IC50 = 2.02 μM). Among these, compounds HD-6, HD-7, and HD-8 displayed the most potent activity with IC50 value of 21.26, 16.75, and 18.33 μM, respectively. Cytotoxicity of all compounds was tested on normal vero cells for comparison at different concentrations using the MTT assay. In addition to the MTT assay, the potent dihydropyridines derivatives were screened for EGFRwt kinase inhibition assay at concentrations ranging from 1 nM to 360 nM. Among the three compounds tested, HD-8 showed reasonably good inhibition with an IC50 value of 15.90 ± 1.20 nM compared to a standard Lapatinib IC50 value of 10.28 ± 1.01 nM. Based on the molecular docking study against EGFR, the most active derivatives HD-7 and HD-8 were docked against the active site of the protein and showed better binding affinity than the standard lapatinib. Additionally, molecular dynamics (MD) simulations were performed to explore the stability of the protein-ligand complex, its dynamic behavior, and the binding affinity.
Collapse
Affiliation(s)
- Syed Faizan
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Constituent College of JSS Academy of Higher Education & Research Mysuru 570015 India +91-821-2548359 +91-821-2548353
| | - Sirajunisa Talath
- Department of Pharmaceutical Chemistry, RAK Medical & Health Sciences University Ras Al Khaimah UAE
| | - Adil Farooq Wali
- Department of Pharmaceutical Chemistry, RAK Medical & Health Sciences University Ras Al Khaimah UAE
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University Abha Saudi Arabia
| | - Nazima Haider
- Department of Pathology, College of Medicine, King Khalid University Abha Saudi Arabia
| | - Subhankar P Mandal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Constituent College of JSS Academy of Higher Education & Research Mysuru 570015 India +91-821-2548359 +91-821-2548353
| | - B R Prashantha Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Constituent College of JSS Academy of Higher Education & Research Mysuru 570015 India +91-821-2548359 +91-821-2548353
| |
Collapse
|
45
|
Khalifa H, Rasheed S, Haupenthal J, Herrmann J, Mandour YM, Abadi AH, Engel M, Müller R, Hirsch AKH, Abdel-Halim M, Hamed MM. Development and evaluation of 2,4-disubstituted-5-aryl pyrimidine derivatives as antibacterial agents. Arch Pharm (Weinheim) 2024; 357:e2300656. [PMID: 38304944 DOI: 10.1002/ardp.202300656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024]
Abstract
Designing novel candidates as potential antibacterial scaffolds has become crucial due to the lack of new antibiotics entering the market and the persistent rise in multidrug resistance. Here, we describe a new class of potent antibacterial agents based on a 5-aryl-N2,N4-dibutylpyrimidine-2,4-diamine scaffold. Structural optimization focused on the 5-aryl moiety and the bioisosteric replacement of the side chain linker atom. Screening of the synthesized compounds focused on a panel of bacterial strains, including gram-positive Staphylococcus aureus strains (Newman MSSA, methicillin- and vancomycin-resistant), and the gram-negative Escherichia coli (ΔAcrB strain). Several compounds showed broad-spectrum antibacterial activity with compound 12, bearing a 4-chlorophenyl substituent, being the most potent among this series of compounds. This frontrunner compound revealed a minimum inhibitory concentration (MIC) value of 1 µg/mL against the S. aureus strain (Mu50 methicillin-resistant S. aureus/vancomycin-intermediate S. aureus) and an MIC of 2 µg/mL against other tested strains. The most potent derivatives were further tested against a wider panel of bacteria and evaluated for their cytotoxicity, revealing further potent activities toward Streptococcus pneumoniae, Enterococcus faecium, and Enterococcus faecalis. To explore the mode of action, compound 12 was tested in a macromolecule inhibition assay. The obtained data were supported by the safety profile of compound 12, which possessed an IC50 of 12.3 µg/mL against HepG2 cells. The current results hold good potential for a new class of extended-spectrum antibacterial agents.
Collapse
Affiliation(s)
- Hend Khalifa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Sari Rasheed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Saarbrucken, Germany
| | - Jörg Haupenthal
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken, Germany
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Saarbrucken, Germany
| | - Yasmine M Mandour
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, Cairo, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbrücken, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Saarbrucken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
- Helmholtz International Lab for Anti-infectives, Saarbrücken, Germany
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Saarbrucken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
- Helmholtz International Lab for Anti-infectives, Saarbrücken, Germany
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Mostafa M Hamed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken, Germany
| |
Collapse
|
46
|
Singh H, Nim DK, Randhawa AS, Ahluwalia S. Integrating clinical pharmacology and artificial intelligence: potential benefits, challenges, and role of clinical pharmacologists. Expert Rev Clin Pharmacol 2024; 17:381-391. [PMID: 38340012 DOI: 10.1080/17512433.2024.2317963] [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: 10/27/2023] [Accepted: 02/08/2024] [Indexed: 02/12/2024]
Abstract
INTRODUCTION The integration of artificial intelligence (AI) into clinical pharmacology could be a potential approach for accelerating drug discovery and development, improving patient care, and streamlining medical research processes. AREAS COVERED We reviewed the current state of AI applications in clinical pharmacology, focusing on drug discovery and development, precision medicine, pharmacovigilance, and other ventures. Key AI applications in clinical pharmacology are examined, including machine learning, natural language processing, deep learning, and reinforcement learning etc. Additionally, the evolving role of clinical pharmacologists, ethical considerations, and challenges in implementing AI in clinical pharmacology are discussed. EXPERT OPINION The AI could be instrumental in accelerating drug discovery, predicting drug safety and efficacy, and optimizing clinical trial designs. It can play a vital role in precision medicine by helping in personalized drug dosing, treatment selection, and predicting drug response based on genetic, clinical, and environmental factors. The role of AI in pharmacovigilance, such as signal detection and adverse event prediction, is also promising. The collaboration between clinical pharmacologists and AI experts also poses certain ethical and practical challenges. Clinical pharmacologists can be instrumental in shaping the future of AI-driven clinical pharmacology and contribute to the improvement of healthcare systems.
Collapse
Affiliation(s)
- Harmanjit Singh
- Department of Pharmacology, Government Medical College & Hospital, Chandigarh, India
| | | | | | | |
Collapse
|
47
|
Humayun F, Khan F, Khan A, Alshammari A, Ji J, Farhan A, Fawad N, Alam W, Ali A, Wei DQ. De novo generation of dual-target ligands for the treatment of SARS-CoV-2 using deep learning, virtual screening, and molecular dynamic simulations. J Biomol Struct Dyn 2024; 42:3019-3029. [PMID: 37449757 DOI: 10.1080/07391102.2023.2234481] [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: 12/26/2022] [Accepted: 04/30/2023] [Indexed: 07/18/2023]
Abstract
De novo generation of molecules with the necessary features offers a promising opportunity for artificial intelligence, such as deep generative approaches. However, creating novel compounds having biological activities toward two distinct targets continues to be a very challenging task. In this study, we develop a unique computational framework for the de novo synthesis of bioactive compounds directed at two predetermined therapeutic targets. This framework is referred to as the dual-target ligand generative network. Our approach uses a stochastic policy to explore chemical spaces called a sequence-based simple molecular input line entry system (SMILES) generator. The steps in the high-level workflow would be to gather and prepare the training data for both targets' molecules, build a neural network model and train it to make molecules, create new molecules using generative AI, and then virtually screen the newly validated molecules against the SARS-CoV-2 PLpro and 3CLpro drug targets. Results shows that novel molecules generated have higher binding affinity with both targets than the conventional drug i.e. Remdesivir being used for the treatment of SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fahad Humayun
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Fatima Khan
- National Institute of Health, Islamabad, Pakistan
| | - Abbas Khan
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Jun Ji
- Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, Henan Provincial Engineering and Technology Center of Animal Disease Diagnosis and Integrated Control, Nanyang Normal University, Nanyang, PR China
| | - Ali Farhan
- Department of Chemistry, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Nasim Fawad
- Poultry Research Institute, Rawalpindi, Pakistan
| | - Waheed Alam
- National Institute of Health, Islamabad, Pakistan
| | - Arif Ali
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Dong-Qing Wei
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
- Centre for Research in Molecular Modeling, Concordia University, Québec, Canada
| |
Collapse
|
48
|
Zhou J, Dong J, Hou H, Huang L, Li J. High-throughput microfluidic systems accelerated by artificial intelligence for biomedical applications. LAB ON A CHIP 2024; 24:1307-1326. [PMID: 38247405 DOI: 10.1039/d3lc01012k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
High-throughput microfluidic systems are widely used in biomedical fields for tasks like disease detection, drug testing, and material discovery. Despite the great advances in automation and throughput, the large amounts of data generated by the high-throughput microfluidic systems generally outpace the abilities of manual analysis. Recently, the convergence of microfluidic systems and artificial intelligence (AI) has been promising in solving the issue by significantly accelerating the process of data analysis as well as improving the capability of intelligent decision. This review offers a comprehensive introduction on AI methods and outlines the current advances of high-throughput microfluidic systems accelerated by AI, covering biomedical detection, drug screening, and automated system control and design. Furthermore, the challenges and opportunities in this field are critically discussed as well.
Collapse
Affiliation(s)
- Jianhua Zhou
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Jianpei Dong
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Hongwei Hou
- Beijing Life Science Academy, Beijing 102209, China
| | - Lu Huang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.
- New Cornerstone Science Laboratory, Shenzhen 518054, China
- Beijing Life Science Academy, Beijing 102209, China
- Center for BioAnalytical Chemistry, Hefei National Laboratory of Physical Science at Microscale, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
49
|
Ai Z, Zhang L, Chen Y, Long Y, Li B, Dong Q, Wang Y, Jiang J. On-Demand Optimization of Colorimetric Gas Sensors Using a Knowledge-Aware Algorithm-Driven Robotic Experimental Platform. ACS Sens 2024; 9:745-752. [PMID: 38331733 DOI: 10.1021/acssensors.3c02043] [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: 02/10/2024]
Abstract
Synthesizing the best material globally is challenging; it needs to know what and how much the best ingredient composition should be for satisfying multiple figures of merit simultaneously. Traditional one-variable-at-a-time methods are inefficient; the design-build-test-learn (DBTL) method could achieve the optimal composition from only a handful of ingredients. A vast design space needs to be explored to discover the possible global optimal composition for on-demand materials synthesis. This research developed a hypothesis-guided DBTL (H-DBTL) method combined with robots to expand the dimensions of the search space, thereby achieving a better global optimal performance. First, this study engineered the search space with knowledge-aware chemical descriptors and customized multiobjective functions to fulfill on-demand research objectives. To verify this concept, this novel method was used to optimize colorimetric ammonia sensors across a vast design space of as high as 19 variables, achieving two remarkable optimization goals within 1 week: first, a sensing array was developed for ammonia quantification of a wide dynamic range, from 0.5 to 500 ppm; second, a new state-of-the-art detection limit of 50 ppb was reached. This work demonstrates that the H-DBTL approach, combined with a robot, develops a novel paradigm for the on-demand optimization of functional materials.
Collapse
Affiliation(s)
- Zhehong Ai
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
- Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
| | - Longhan Zhang
- Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
| | - Yangguan Chen
- Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
| | - Yifan Long
- Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
| | - Boyuan Li
- Hong Kong Center for Construction Robotics Limited, Hong Kong 808-815, China
| | - Qingyu Dong
- Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
- Polytechnic Institute, Zhejiang University, Hangzhou, Zhejiang 310015, China
| | - Yueming Wang
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
- Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Space Active Optoelectronics Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, China
| | - Jing Jiang
- Zhejiang Laboratory, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
50
|
Yang S, Kar S. Protracted molecular dynamics and secondary structure introspection to identify dual-target inhibitors of Nipah virus exerting approved small molecules repurposing. Sci Rep 2024; 14:3696. [PMID: 38355980 PMCID: PMC10866979 DOI: 10.1038/s41598-024-54281-9] [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: 12/01/2023] [Accepted: 02/10/2024] [Indexed: 02/16/2024] Open
Abstract
Nipah virus (NiV), with its significantly higher mortality rate compared to COVID-19, presents a looming threat as a potential next pandemic, particularly if constant mutations of NiV increase its transmissibility and transmission. Considering the importance of preventing the facilitation of the virus entry into host cells averting the process of assembly forming the viral envelope, and encapsulating the nucleocapsid, it is crucial to take the Nipah attachment glycoprotein-human ephrin-B2 and matrix protein as dual targets. Repurposing approved small molecules in drug development is a strategic choice, as it leverages molecules with known safety profiles, accelerating the path to finding effective treatments against NiV. The approved small molecules from DrugBank were used for repurposing and were subjected to extra precision docking followed by absorption, distribution, metabolism, excretion, and toxicity (ADMET) profiling. The 4 best molecules were selected for 500 ns molecular dynamics (MD) simulation followed by Molecular mechanics with generalized Born and surface area solvation (MM-GBSA). Further, the free energy landscape, the principal component analysis followed by the defined secondary structure of proteins analysis were introspected. The inclusive analysis proposed that Iotrolan (DB09487) and Iodixanol (DB01249) are effective dual inhibitors, while Rutin (DB01698) and Lactitol (DB12942) were found to actively target the matrix protein only.
Collapse
Affiliation(s)
- Siyun Yang
- Chemometrics and Molecular Modeling Laboratory, Department of Chemistry and Physics, Kean University, 1000 Morris Avenue, Union, NJ, 07083, USA
| | - Supratik Kar
- Chemometrics and Molecular Modeling Laboratory, Department of Chemistry and Physics, Kean University, 1000 Morris Avenue, Union, NJ, 07083, USA.
| |
Collapse
|