1
|
Li Z, Ruan Q, Jiang Y, Wang Q, Yin G, Feng J, Zhang J. Current Status and Perspectives of Novel Radiopharmaceuticals with Heterologous Dual-targeted Functions: 2013-2023. J Med Chem 2024; 67:21644-21670. [PMID: 39648432 DOI: 10.1021/acs.jmedchem.4c01608] [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: 12/10/2024]
Abstract
Radiotracers provide molecular- and cellular-level information in a noninvasive manner and have become important tools for precision medicine. In particular, the successful clinical application of radioligand therapeutic (RLT) has further strengthened the role of nuclear medicine in clinical treatment. The complicated microenvironment of the lesion has rendered traditional single-targeted radiopharmaceuticals incapable of fully meeting the requirements. The design and development of dual-targeted and multitargeted radiopharmaceuticals have rapidly emerged. In recent years, significant progress has been made in the development of heterologous dual-targeted radiopharmaceuticals. This perspective aims to provide a comprehensive overview of the recent progress in these heterologous dual-targeted radiopharmaceuticals, with a special focus on the design of ligand structures, pharmacological properties, and preclinical and clinical evaluation. Furthermore, future directions are discussed from this perspective.
Collapse
Affiliation(s)
- Zuojie Li
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Qing Ruan
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- Key Laboratory of Beam Technology of the Ministry of Education, College of Physics and Astronomy, Beijing Normal University, Beijing, 100875, P. R. China
| | - Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- Key Laboratory of Beam Technology of the Ministry of Education, College of Physics and Astronomy, Beijing Normal University, Beijing, 100875, P. R. China
| | - Qianna Wang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Guangxing Yin
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Junhong Feng
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| |
Collapse
|
2
|
Ambreen S, Umar M, Noor A, Jain H, Ali R. Advanced AI and ML frameworks for transforming drug discovery and optimization: With innovative insights in polypharmacology, drug repurposing, combination therapy and nanomedicine. Eur J Med Chem 2024; 284:117164. [PMID: 39721292 DOI: 10.1016/j.ejmech.2024.117164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024]
Abstract
Artificial Intelligence (AI) and Machine Learning (ML) are transforming drug discovery by overcoming traditional challenges like high costs, time-consuming, and frequent failures. AI-driven approaches streamline key phases, including target identification, lead optimization, de novo drug design, and drug repurposing. Frameworks such as deep neural networks (DNNs), convolutional neural networks (CNNs), and deep reinforcement learning (DRL) models have shown promise in identifying drug targets, optimizing delivery systems, and accelerating drug repurposing. Generative adversarial networks (GANs) and variational autoencoders (VAEs) aid de novo drug design by creating novel drug-like compounds with desired properties. Case studies, such as DDR1 kinase inhibitors designed using generative models and CDK20 inhibitors developed via structure-based methods, highlight AI's ability to produce highly specific therapeutics. Models like SNF-CVAE and DeepDR further advance drug repurposing by uncovering new therapeutic applications for existing drugs. Advanced ML algorithms enhance precision in predicting drug efficacy, toxicity, and ADME-Tox properties, reducing development costs and improving drug-target interactions. AI also supports polypharmacology by optimizing multi-target drug interactions and enhances combination therapy through predictions of drug synergies and antagonisms. In nanomedicine, AI models like CURATE.AI and the Hartung algorithm optimize personalized treatments by predicting toxicological risks and real-time dosing adjustments with high accuracy. Despite its potential, challenges like data quality, model interpretability, and ethical concerns must be addressed. High-quality datasets, transparent models, and unbiased algorithms are essential for reliable AI applications. As AI continues to evolve, it is poised to revolutionize drug discovery and personalized medicine, advancing therapeutic development and patient care.
Collapse
Affiliation(s)
- Subiya Ambreen
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Mohammad Umar
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Aaisha Noor
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Himangini Jain
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Ruhi Ali
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India.
| |
Collapse
|
3
|
Yang X, Duan Y, Cheng Z, Li K, Liu Y, Zeng X, Cao D. MPCD: A Multitask Graph Transformer for Molecular Property Prediction by Integrating Common and Domain Knowledge. J Med Chem 2024; 67:21303-21316. [PMID: 39620982 DOI: 10.1021/acs.jmedchem.4c02193] [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: 12/13/2024]
Abstract
Molecular property prediction with deep learning often employs self-supervised learning techniques to learn common knowledge through masked atom prediction. However, the common knowledge gained by masked atom prediction dramatically differs from the graph-level optimization objective of downstream tasks, which results in suboptimal problems. Particularly for properties with limited data, the failure to consider domain knowledge results in a direct search in an immense common space, rendering it infeasible to identify the global optimum. To address this, we propose MPCD, which enhances pretraining transferability by aligning the optimization objectives between pretraining and fine-tuning with domain knowledge. MPCD also leverages multitask learning to improve data utilization and model robustness. Technically, MPCD employs a relation-aware self-attention mechanism to capture molecules' local and global structures comprehensively. Extensive validation demonstrates that MPCD outperforms state-of-the-art methods for absorption, distribution, metabolism, excretion, and toxicity (ADMET) and physicochemical prediction across various data sizes.
Collapse
Affiliation(s)
- Xixi Yang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410086, Hunan, China
| | - Yanjing Duan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Zhixiang Cheng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410086, Hunan, China
| | - Kun Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Yuansheng Liu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410086, Hunan, China
| | - Xiangxiang Zeng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410086, Hunan, China
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| |
Collapse
|
4
|
Mohammed I, Sagurthi SR. Current Approaches and Strategies Applied in First-in-class Drug Discovery. ChemMedChem 2024:e202400639. [PMID: 39648151 DOI: 10.1002/cmdc.202400639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
First-in-class drug discovery (FICDD) offers novel therapies, new biological targets and mechanisms of action (MOAs) toward targeting various diseases and provides opportunities to understand unexplored biology and to target unmet diseases. Current screening approaches followed in FICDD for discovery of hit and lead molecules can be broadly categorized and discussed under phenotypic drug discovery (PDD) and target-based drug discovery (TBDD). Each category has been further classified and described with suitable examples from the literature outlining the current trends in screening approaches applied in small molecule drug discovery (SMDD). Similarly, recent applications of functional genomics, structural biology, artificial intelligence (AI), machine learning (ML), and other such advanced approaches in FICDD have also been highlighted in the article. Further, some of the current medicinal chemistry strategies applied during discovery of hits and optimization studies such as hit-to-lead (HTL) and lead optimization (LO) have been simultaneously overviewed in this article.
Collapse
Affiliation(s)
- Idrees Mohammed
- Drug Design & Molecular Medicine Laboratory, Department of Genetics & Biotechnology, Osmania University, Hyderabad, 500007, Telangana, India
| | - Someswar Rao Sagurthi
- Drug Design & Molecular Medicine Laboratory, Department of Genetics & Biotechnology, Osmania University, Hyderabad, 500007, Telangana, India
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
5
|
Zhang Y, Cai J, Hosmane NS, Zhu Y. In silico assessments of the small molecular boron agents to pave the way for artificial intelligence-based boron neutron capture therapy. Eur J Med Chem 2024; 279:116841. [PMID: 39244862 DOI: 10.1016/j.ejmech.2024.116841] [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: 03/21/2024] [Revised: 07/28/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024]
Abstract
Boron neutron capture therapy (BNCT) is a highly targeted, selective and effective technique to cure various types of cancers, with less harm to the healthy cells. In principle, BNCT treatment needs to distribute the 10boron (10B) atoms inside the tumor tissues, selectively and homogeneously, as well as to initiate a nuclear fission reaction by capturing sufficient neutrons which releases high linear energy particles to kill the tumor cells. In BNCT, it is crucial to have high quality boron agents with acceptable bio-selectivity, homogeneous distribution and deliver in required quantity, similar to chemotherapy and other radiotherapy for tumor treatment. Nevertheless, boron drugs currently used in clinical trials yet to meet the full requirements. On the other hand, BNCT processing has opened up the era of renaissance due to the advanced development of the high-quality neutron source and the global construction of new BNCT centers. Consequently, there is an urgent need to use boron agents that have increased biocapacity. Artificial intelligence (AI) tools such as molecular docking and molecular dynamic simulation technologies have been utilized to develop new medicines. In this work, the in silico assessments including bioinformatics assessments of BNCT related tumoral receptor proteins, computational assessments of optimized small molecules of boron agents, are employed to speed up the screening process for boron drugs. The outcomes will be applicable to pave the way for future BNCT that utilizes artificial intelligence. The in silico molecular docking and dynamic simulation results of the optimized small boron agents, such as 4-borono-l-phenylalanine (BPA) with optimized proteins like the L-type amino acid transporter 1 (LTA1, also known as SLC7A5) will be examined. The in silico assessments results will certainly be helpful to researchers in optimizing druggable boron agents for the BNCT application. The clinical status of the optimized proteins, which are highly relevant to cancers that may be treated with BNCT, has been assessed using bioinformatics technology and discussed accordingly. Furthermore, the evaluations of cytotoxicity (IC50), boron uptake and tissue distribution of the optimized ligands 1 and 7 have been presented.
Collapse
Affiliation(s)
- Yingjun Zhang
- Sunshine Lake Pharma Co. Ltd., Dongguan, 523871, China
| | - Jianghong Cai
- School of Pharmacy, Macau University of Science and Technology, Macau, 999078, China
| | - Narayan S Hosmane
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, IL, USA
| | - Yinghuai Zhu
- Sunshine Lake Pharma Co. Ltd., Dongguan, 523871, China.
| |
Collapse
|
6
|
Campos TL, Korhonen PK, Young ND, Chang BC, Gasser RB. Inference of essential genes in Brugia malayi and Onchocerca volvulus by machine learning and the implications for discovering new interventions. Comput Struct Biotechnol J 2024; 23:3081-3089. [PMID: 39185442 PMCID: PMC11342751 DOI: 10.1016/j.csbj.2024.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/27/2024] Open
Abstract
Detailed explorations of the model organisms Caenorhabditis elegans (elegant worm) and Drosophila melanogaster (vinegar fly) have substantially improved our knowledge and understanding of biological processes and pathways in metazoan organisms. Extensive functional genomic and multi-omic data sets have enabled the discovery and characterisation of 'essential' genes that are critical for the survival of these organisms. Recently, we showed that a machine learning (ML)-based pipeline could be utilised to predict essential genes in both C. elegans and D. melanogaster using features from DNA, RNA, protein and/or cellular data or associated information. As these distantly-related species are within the Ecdysozoa, we hypothesised that this approach could be suited for non-model organisms within the same group (phylum) of protostome animals. In the present investigation, we cross-predicted essential genes within the phylum Nematoda - between C. elegans and the parasitic filarial nematodes Brugia malayi and Onchocerca volvulus, and then ranked and prioritised these genes. Highly ranked genes were linked to key biological pathways or processes, such as ribosome biogenesis, translation and RNA processing, and were expressed at relatively high levels in the germline, gonad, hypodermis and/or nerves. The present in silico workflow is hoped to expedite the identification of drug targets in parasitic organisms for subsequent experimental validation in the laboratory.
Collapse
Affiliation(s)
- Túlio L. Campos
- Department of Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
- Núcleo de Bioinformática, Instituto Aggeu Magalhães, Fiocruz., Av. Professor Moraes Rego, s/n, Cidade Universitária, Recife, PE CEP 50740–465, Brazil
| | - Pasi K. Korhonen
- Department of Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Neil D. Young
- Department of Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Bill C.H. Chang
- Department of Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Robin B. Gasser
- Department of Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
7
|
Gao J, Wang D. Quantifying the use and potential benefits of artificial intelligence in scientific research. Nat Hum Behav 2024; 8:2281-2292. [PMID: 39394445 DOI: 10.1038/s41562-024-02020-5] [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: 04/26/2023] [Accepted: 09/12/2024] [Indexed: 10/13/2024]
Abstract
The rapid advancement of artificial intelligence (AI) is poised to reshape almost every line of work. Despite enormous efforts devoted to understanding AI's economic impacts, we lack a systematic understanding of the benefits to scientific research associated with the use of AI. Here we develop a measurement framework to estimate the direct use of AI and associated benefits in science. We find that the use and benefits of AI appear widespread throughout the sciences, growing especially rapidly since 2015. However, there is a substantial gap between AI education and its application in research, highlighting a misalignment between AI expertise supply and demand. Our analysis also reveals demographic disparities, with disciplines with higher proportions of women or Black scientists reaping fewer benefits from AI, potentially exacerbating existing inequalities in science. These findings have implications for the equity and sustainability of the research enterprise, especially as the integration of AI with science continues to deepen.
Collapse
Affiliation(s)
- Jian Gao
- Center for Science of Science and Innovation, Northwestern University, Evanston, IL, USA
- Kellogg School of Management, Northwestern University, Evanston, IL, USA
- Ryan Institute on Complexity, Northwestern University, Evanston, IL, USA
- Faculty of Social Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Dashun Wang
- Center for Science of Science and Innovation, Northwestern University, Evanston, IL, USA.
- Kellogg School of Management, Northwestern University, Evanston, IL, USA.
- Ryan Institute on Complexity, Northwestern University, Evanston, IL, USA.
- McCormick School of Engineering, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
8
|
Jones D, Zhang X, Bennion BJ, Pinge S, Xu W, Kang J, Khaleghi B, Moshiri N, Allen JE, Rosing TS. HDBind: encoding of molecular structure with hyperdimensional binary representations. Sci Rep 2024; 14:29025. [PMID: 39578580 PMCID: PMC11584749 DOI: 10.1038/s41598-024-80009-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024] Open
Abstract
Traditional methods for identifying "hit" molecules from a large collection of potential drug-like candidates rely on biophysical theory to compute approximations to the Gibbs free energy of the binding interaction between the drug and its protein target. These approaches have a significant limitation in that they require exceptional computing capabilities for even relatively small collections of molecules. Increasingly large and complex state-of-the-art deep learning approaches have gained popularity with the promise to improve the productivity of drug design, notorious for its numerous failures. However, as deep learning models increase in their size and complexity, their acceleration at the hardware level becomes more challenging. Hyperdimensional Computing (HDC) has recently gained attention in the computer hardware community due to its algorithmic simplicity relative to deep learning approaches. The HDC learning paradigm, which represents data with high-dimension binary vectors, allows the use of low-precision binary vector arithmetic to create models of the data that can be learned without the need for the gradient-based optimization required in many conventional machine learning and deep learning methods. This algorithmic simplicity allows for acceleration in hardware that has been previously demonstrated in a range of application areas (computer vision, bioinformatics, mass spectrometery, remote sensing, edge devices, etc.). To the best of our knowledge, our work is the first to consider HDC for the task of fast and efficient screening of modern drug-like compound libraries. We also propose the first HDC graph-based encoding methods for molecular data, demonstrating consistent and substantial improvement over previous work. We compare our approaches to alternative approaches on the well-studied MoleculeNet dataset and the recently proposed LIT-PCBA dataset derived from high quality PubChem assays. We demonstrate our methods on multiple target hardware platforms, including Graphics Processing Units (GPUs) and Field Programmable Gate Arrays (FPGAs), showing at least an order of magnitude improvement in energy efficiency versus even our smallest neural network baseline model with a single hidden layer. Our work thus motivates further investigation into molecular representation learning to develop ultra-efficient pre-screening tools. We make our code publicly available at https://github.com/LLNL/hdbind .
Collapse
Affiliation(s)
- Derek Jones
- Department of Computer Science and Engineering, University of California-San Diego, La Jolla, CA, USA.
- Global Security Computing Applications Division, Lawrence Livermore National Laboratory, Livermore, CA, USA.
| | - Xiaohua Zhang
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Brian J Bennion
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Sumukh Pinge
- Department of Computer Science and Engineering, University of California-San Diego, La Jolla, CA, USA
| | - Weihong Xu
- Department of Computer Science and Engineering, University of California-San Diego, La Jolla, CA, USA
| | - Jaeyoung Kang
- Department of Computer Science and Engineering, University of California-San Diego, La Jolla, CA, USA
| | - Behnam Khaleghi
- Department of Computer Science and Engineering, University of California-San Diego, La Jolla, CA, USA
| | - Niema Moshiri
- Department of Computer Science and Engineering, University of California-San Diego, La Jolla, CA, USA
| | - Jonathan E Allen
- Global Security Computing Applications Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Tajana S Rosing
- Department of Computer Science and Engineering, University of California-San Diego, La Jolla, CA, USA
| |
Collapse
|
9
|
Flores-Hernandez H, Martinez-Ledesma E. A systematic review of deep learning chemical language models in recent era. J Cheminform 2024; 16:129. [PMID: 39558376 PMCID: PMC11571686 DOI: 10.1186/s13321-024-00916-y] [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: 07/26/2024] [Accepted: 10/17/2024] [Indexed: 11/20/2024] Open
Abstract
Discovering new chemical compounds with specific properties can provide advantages for fields that rely on materials for their development, although this task comes at a high cost in terms of complexity and resources. Since the beginning of the data age, deep learning techniques have revolutionized the process of designing molecules by analyzing and learning from representations of molecular data, greatly reducing the resources and time involved. Various deep learning approaches have been developed to date, using a variety of architectures and strategies, in order to explore the extensive and discontinuous chemical space, providing benefits for generating compounds with specific properties. In this study, we present a systematic review that offers a statistical description and comparison of the strategies utilized to generate molecules through deep learning techniques, utilizing the metrics proposed in Molecular Sets (MOSES) or Guacamol. The study included 48 articles retrieved from a query-based search of Scopus and Web of Science and 25 articles retrieved from citation search, yielding a total of 72 retrieved articles, of which 62 correspond to chemical language models approaches to molecule generation and other 10 retrieved articles correspond to molecular graph representations. Transformers, recurrent neural networks (RNNs), generative adversarial networks (GANs), Structured Space State Sequence (S4) models, and variational autoencoders (VAEs) are considered the main deep learning architectures used for molecule generation in the set of retrieved articles. In addition, transfer learning, reinforcement learning, and conditional learning are the most employed techniques for biased model generation and exploration of specific chemical space regions. Finally, this analysis focuses on the central themes of molecular representation, databases, training dataset size, validity-novelty trade-off, and performance of unbiased and biased chemical language models. These themes were selected to conduct a statistical analysis utilizing graphical representation and statistical tests. The resulting analysis reveals the main challenges, advantages, and opportunities in the field of chemical language models over the past four years.
Collapse
Affiliation(s)
- Hector Flores-Hernandez
- Tecnológico de Monterrey, School of Engineering and Sciences, Monterrey, 64710, Nuevo León, México
| | - Emmanuel Martinez-Ledesma
- Tecnológico de Monterrey, School of Medicine and Health Sciences, Monterrey, 64710, Nuevo León, México.
- Institute for Obesity Research, Tecnológico de Monterrey, Monterrey, 64710, Nuevo León, México.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Yang Y, Mou Y, Wan LX, Zhu S, Wang G, Gao H, Liu B. Rethinking therapeutic strategies of dual-target drugs: An update on pharmacological small-molecule compounds in cancer. Med Res Rev 2024; 44:2600-2623. [PMID: 38769656 DOI: 10.1002/med.22057] [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: 02/15/2022] [Revised: 06/06/2023] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Oncogenes and tumor suppressors are well-known to orchestrate several signaling cascades, regulate extracellular and intracellular stimuli, and ultimately control the fate of cancer cells. Accumulating evidence has recently revealed that perturbation of these key modulators by mutations or abnormal protein expressions are closely associated with drug resistance in cancer therapy; however, the inherent drug resistance or compensatory mechanism remains to be clarified for targeted drug discovery. Thus, dual-target drug development has been widely reported to be a promising therapeutic strategy for improving drug efficiency or overcoming resistance mechanisms. In this review, we provide an overview of the therapeutic strategies of dual-target drugs, especially focusing on pharmacological small-molecule compounds in cancer, including small molecules targeting mutation resistance, compensatory mechanisms, synthetic lethality, synergistic effects, and other new emerging strategies. Together, these therapeutic strategies of dual-target drugs would shed light on discovering more novel candidate small-molecule drugs for the future cancer treatment.
Collapse
Affiliation(s)
- Yiren Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yi Mou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Lin-Xi Wan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Shiou Zhu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Guan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Huiyuan Gao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Bo Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Sharma R, Oyagawa CRM, Abbasi H, Dragunow M, Conole D. Phenotypic approaches for CNS drugs. Trends Pharmacol Sci 2024; 45:997-1017. [PMID: 39438155 DOI: 10.1016/j.tips.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/09/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Central nervous system (CNS) drug development is plagued by high clinical failure rate. Phenotypic assays promote clinical translation of drugs by reducing complex brain diseases to measurable, clinically valid phenotypes. We critique recent platforms integrating patient-derived brain cells, which most accurately recapitulate CNS disease phenotypes, with higher throughput models, including immortalized cells, to balance validity and scalability. These platforms were screened with conventional commercial chemogenomic compound libraries. We explore emerging library curation strategies to improve hit rate and quality, and screening novel fragment libraries as alternatives, for more tractable drug target deconvolution. The clinically relevant models used in these platforms could harbor important, unidentified drug targets, so we review evolving agnostic target deconvolution approaches, including chemical proteomics and artificial intelligence (AI), which aid in phenotypic screening hit mechanism elucidation, thereby facilitating rational hit-to-drug optimization.
Collapse
Affiliation(s)
- Raahul Sharma
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand; Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand
| | - Caitlin R M Oyagawa
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand
| | - Hamid Abbasi
- Auckland Bioengineering Institute, The University of Auckland, 70 Symonds Street, Auckland, 1010, New Zealand
| | - Michael Dragunow
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand.
| | - Daniel Conole
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand.
| |
Collapse
|
14
|
Mao X, Chu Y, Wei D. Designed with interactome-based deep learning. Nat Chem Biol 2024; 20:1399-1401. [PMID: 39424957 DOI: 10.1038/s41589-024-01754-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Affiliation(s)
- Xueying Mao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Yanyi Chu
- Department of Electrical and Computer Engineering, Princeton University, Princeton, NJ, USA
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Dongqing Wei
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, P.R. China.
- Qihe Laboratory, Qibin District, Hebi, P.R. China.
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Nanyang, P.R. China.
- Peng Cheng National Laboratory, Shenzhen, P.R. China.
| |
Collapse
|
15
|
Wu K, Xia Y, Deng P, Liu R, Zhang Y, Guo H, Cui Y, Pei Q, Wu L, Xie S, Chen S, Lu X, Hu S, Wu J, Chan CK, Chen S, Zhou L, Yu N, Chen E, Liu H, Guo J, Qin T, Liu TY. TamGen: drug design with target-aware molecule generation through a chemical language model. Nat Commun 2024; 15:9360. [PMID: 39472567 PMCID: PMC11522292 DOI: 10.1038/s41467-024-53632-4] [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: 03/15/2024] [Accepted: 10/14/2024] [Indexed: 11/02/2024] Open
Abstract
Generative drug design facilitates the creation of compounds effective against pathogenic target proteins. This opens up the potential to discover novel compounds within the vast chemical space and fosters the development of innovative therapeutic strategies. However, the practicality of generated molecules is often limited, as many designs focus on a narrow set of drug-related properties, failing to improve the success rate of subsequent drug discovery process. To overcome these challenges, we develop TamGen, a method that employs a GPT-like chemical language model and enables target-aware molecule generation and compound refinement. We demonstrate that the compounds generated by TamGen have improved molecular quality and viability. Additionally, we have integrated TamGen into a drug discovery pipeline and identified 14 compounds showing compelling inhibitory activity against the Tuberculosis ClpP protease, with the most effective compound exhibiting a half maximal inhibitory concentration (IC50) of 1.9 μM. Our findings underscore the practical potential and real-world applicability of generative drug design approaches, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Kehan Wu
- University of Science and Technology of China, Hefei, China
| | - Yingce Xia
- Microsoft Research AI for Science, Beijing, China.
| | - Pan Deng
- Microsoft Research AI for Science, Beijing, China
| | - Renhe Liu
- Global Health Drug Discovery Institute, Beijing, China
| | - Yuan Zhang
- Global Health Drug Discovery Institute, Beijing, China
| | - Han Guo
- Global Health Drug Discovery Institute, Beijing, China
| | - Yumeng Cui
- Global Health Drug Discovery Institute, Beijing, China
| | - Qizhi Pei
- Renmin University of China, Beijing, China
| | - Lijun Wu
- Microsoft Research AI for Science, Beijing, China
| | - Shufang Xie
- Microsoft Research AI for Science, Beijing, China
| | - Si Chen
- Global Health Drug Discovery Institute, Beijing, China
| | - Xi Lu
- Global Health Drug Discovery Institute, Beijing, China
| | - Song Hu
- Global Health Drug Discovery Institute, Beijing, China
| | - Jinzhi Wu
- Global Health Drug Discovery Institute, Beijing, China
| | - Chi-Kin Chan
- Global Health Drug Discovery Institute, Beijing, China
| | - Shawn Chen
- Global Health Drug Discovery Institute, Beijing, China
| | | | - Nenghai Yu
- University of Science and Technology of China, Hefei, China
| | - Enhong Chen
- University of Science and Technology of China, Hefei, China
| | - Haiguang Liu
- Microsoft Research AI for Science, Beijing, China
| | - Jinjiang Guo
- Global Health Drug Discovery Institute, Beijing, China.
| | - Tao Qin
- Microsoft Research AI for Science, Beijing, China.
| | - Tie-Yan Liu
- Microsoft Research AI for Science, Beijing, China
| |
Collapse
|
16
|
Asfa SS, Arshinchi Bonab R, Önder O, Uça Apaydın M, Döşeme H, Küçük C, Georgakilas AG, Stadler BM, Logotheti S, Kale S, Pavlopoulou A. Computer-Aided Identification and Design of Ligands for Multi-Targeting Inhibition of a Molecular Acute Myeloid Leukemia Network. Cancers (Basel) 2024; 16:3607. [PMID: 39518047 PMCID: PMC11544916 DOI: 10.3390/cancers16213607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Acute myeloid leukemia (AML) is characterized by therapeutic failure and long-term risk for disease relapses. As several therapeutic targets participate in networks, they can rewire to eventually evade single-target drugs. Hence, multi-targeting approaches are considered on the expectation that interference with many different components could synergistically hinder activation of alternative pathways and demolish the network one-off, leading to complete disease remission. METHODS Herein, we established a network-based, computer-aided approach for the rational design of drug combinations and de novo agents that interact with many AML network components simultaneously. RESULTS A reconstructed AML network guided the selection of suitable protein hubs and corresponding multi-targeting strategies. For proteins responsive to existing drugs, a greedy algorithm identified the minimum amount of compounds targeting the maximum number of hubs. We predicted permissible combinations of amiodarone, artenimol, fostamatinib, ponatinib, procaine, and vismodegib that interfere with 3-8 hubs, and we elucidated the pharmacological mode of action of procaine on DNMT3A. For proteins that do not respond to any approved drugs, namely cyclins A1, D2, and E1, we used structure-based de novo drug design to generate a novel triple-targeting compound of the chemical formula C15H15NO5, with favorable pharmacological and drug-like properties. CONCLUSIONS Overall, by integrating network and structural pharmacology with molecular modeling, we determined two complementary strategies with the potential to annihilate the AML network, one in the form of repurposable drug combinations and the other as a de novo synthesized triple-targeting agent. These target-drug interactions could be prioritized for preclinical and clinical testing toward precision medicine for AML.
Collapse
Affiliation(s)
- Seyedeh Sadaf Asfa
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
- Department of Pharmacology and Therapeutics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R3E 0W2, Canada
| | - Reza Arshinchi Bonab
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
- Department of Pharmacology and Therapeutics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R3E 0W2, Canada
| | - Onur Önder
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
| | - Merve Uça Apaydın
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
| | - Hatice Döşeme
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
| | - Can Küçük
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylül University, 35330 Balçova, İzmir, Türkiye;
| | - Alexandros G. Georgakilas
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou Campous, 15780 Athens, Greece;
| | - Bernhard M. Stadler
- Technische Hochschule Nürnberg, Faculty of Applied Chemistry, 90489 Nuremberg, Germany;
| | - Stella Logotheti
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Seyit Kale
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Department of Biophysics, Faculty of Medicine, Izmir Katip Çelebi University, 35330 Çiğli, İzmir, Türkiye
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
| |
Collapse
|
17
|
Ates Ö, Pandey G, Gousiopoulos A, Soldatos TG. A brief reference to AI-driven audible reality (AuRa) in open world: potential, applications, and evaluation. Front Artif Intell 2024; 7:1424371. [PMID: 39525498 PMCID: PMC11543578 DOI: 10.3389/frai.2024.1424371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024] Open
Abstract
Recent developments on artificial intelligence (AI) and machine learning (ML) techniques are expected to have significant impact on public health in several ways. Indeed, modern AI/ML methods have been applied on multiple occasions on topics ranging from drug discovery and disease diagnostics to personalized medicine, medical imaging, and healthcare operations. While such developments may improve several quality-of-life aspects (such as access to health services and education), it is important considering that some individuals may face more challenges, particularly in extreme or emergency situations. In this work, we focus on utilizing AI/ML components to support scenarios when visual impairment or other limitations hinder the ability to interpret the world in this way. Specifically, we discuss the potential and the feasibility of automatically transferring key visual information into audio communication, in different languages and in real-time-a setting which we name 'audible reality' (AuRa). We provide a short guide to practical options currently available for implementing similar solutions and summarize key aspects for evaluating their scope. Finally, we discuss diverse settings and functionalities that AuRA applications could have in terms of broader impact, from a social and public health context, and invite the community to further such digital solutions and perspectives soon.
Collapse
Affiliation(s)
- Ömer Ates
- School of Information, Media, and Design, SRH Hochschule Heidelberg, SRH University of Applied Science, Heidelberg, Germany
| | - Garima Pandey
- School of Information, Media, and Design, SRH Hochschule Heidelberg, SRH University of Applied Science, Heidelberg, Germany
| | - Athanasios Gousiopoulos
- Department of Library, Archives and Information Systems, School of Social Sciences, International Hellenic University, Thessaloniki, Greece
- Department of Accounting and Information Systems, School of Economics and Business Administration, International Hellenic University, Thessaloniki, Greece
| | - Theodoros G. Soldatos
- School of Information, Media, and Design, SRH Hochschule Heidelberg, SRH University of Applied Science, Heidelberg, Germany
| |
Collapse
|
18
|
Serrano DR, Luciano FC, Anaya BJ, Ongoren B, Kara A, Molina G, Ramirez BI, Sánchez-Guirales SA, Simon JA, Tomietto G, Rapti C, Ruiz HK, Rawat S, Kumar D, Lalatsa A. Artificial Intelligence (AI) Applications in Drug Discovery and Drug Delivery: Revolutionizing Personalized Medicine. Pharmaceutics 2024; 16:1328. [PMID: 39458657 PMCID: PMC11510778 DOI: 10.3390/pharmaceutics16101328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Artificial intelligence (AI) encompasses a broad spectrum of techniques that have been utilized by pharmaceutical companies for decades, including machine learning, deep learning, and other advanced computational methods. These innovations have unlocked unprecedented opportunities for the acceleration of drug discovery and delivery, the optimization of treatment regimens, and the improvement of patient outcomes. AI is swiftly transforming the pharmaceutical industry, revolutionizing everything from drug development and discovery to personalized medicine, including target identification and validation, selection of excipients, prediction of the synthetic route, supply chain optimization, monitoring during continuous manufacturing processes, or predictive maintenance, among others. While the integration of AI promises to enhance efficiency, reduce costs, and improve both medicines and patient health, it also raises important questions from a regulatory point of view. In this review article, we will present a comprehensive overview of AI's applications in the pharmaceutical industry, covering areas such as drug discovery, target optimization, personalized medicine, drug safety, and more. By analyzing current research trends and case studies, we aim to shed light on AI's transformative impact on the pharmaceutical industry and its broader implications for healthcare.
Collapse
Affiliation(s)
- Dolores R. Serrano
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
- Instituto Universitario de Farmacia Industrial, 28040 Madrid, Spain
| | - Francis C. Luciano
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Brayan J. Anaya
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Baris Ongoren
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Aytug Kara
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Gracia Molina
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Bianca I. Ramirez
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Sergio A. Sánchez-Guirales
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Jesus A. Simon
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Greta Tomietto
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Chrysi Rapti
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Helga K. Ruiz
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Satyavati Rawat
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India; (S.R.); (D.K.)
| | - Dinesh Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India; (S.R.); (D.K.)
| | - Aikaterini Lalatsa
- Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161, Cathedral Street, Glasgow G4 0RE, UK
- CRUK Formulation Unit, School of Pharmacy and Biomedical Sciences, University of Strathclyde, 161, Cathedral Street, Glasgow G4 0RE, UK
| |
Collapse
|
19
|
Wang K, Huang Y, Wang Y, You Q, Wang L. Recent advances from computer-aided drug design to artificial intelligence drug design. RSC Med Chem 2024; 15:d4md00522h. [PMID: 39493228 PMCID: PMC11523840 DOI: 10.1039/d4md00522h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
Computer-aided drug design (CADD), a cornerstone of modern drug discovery, can predict how a molecular structure relates to its activity and interacts with its target using structure-based and ligand-based methods. Fueled by ever-increasing data availability and continuous model optimization, artificial intelligence drug design (AIDD), as an enhanced iteration of CADD, has thrived in the past decade. AIDD demonstrates unprecedented opportunities in protein folding, property prediction, and molecular generation. It can also facilitate target identification, high-throughput screening (HTS), and synthetic route prediction. With AIDD involved, the process of drug discovery is greatly accelerated. Notably, AIDD offers the potential to explore uncharted territories of chemical space beyond current knowledge. In this perspective, we began by briefly outlining the main workflows and components of CADD. Then through showcasing exemplary cases driven by AIDD in recent years, we describe the evolving role of artificial intelligence (AI) in drug discovery from three distinct stages, that is, chemical library screening, linker generation, and de novo molecular generation. In this process, we attempted to draw comparisons between the features of CADD and AIDD.
Collapse
Affiliation(s)
- Keran Wang
- State Key Laboratory of Natural Medicines and, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University Nanjing 210009 China +86 025 83271351 +86 15261483858
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University Nanjing 210009 China
| | - Yanwen Huang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University Beijing 100191 China
| | - Yan Wang
- Department of Urology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Shanghai 201203 China +86 13122152007
| | - Qidong You
- State Key Laboratory of Natural Medicines and, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University Nanjing 210009 China +86 025 83271351 +86 15261483858
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University Nanjing 210009 China
| | - Lei Wang
- State Key Laboratory of Natural Medicines and, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University Nanjing 210009 China +86 025 83271351 +86 15261483858
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University Nanjing 210009 China
| |
Collapse
|
20
|
Zaaijer S, Groen SC. Implementing differentially pigmented skin models for predicting drug response variability across human ancestries. Hum Genomics 2024; 18:113. [PMID: 39385300 PMCID: PMC11465898 DOI: 10.1186/s40246-024-00677-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/21/2024] [Indexed: 10/12/2024] Open
Abstract
Persistent racial disparities in health outcomes have catalyzed legislative reforms and heightened scientific focus recently. However, despite the well-documented properties of skin pigments in binding drug compounds, their impact on therapeutic efficacy and adverse drug responses remains insufficiently explored. This perspective examines the intricate relationships between variation in melanin-based skin pigmentation and pharmacokinetics and -dynamics, highlighting the need for considering diversity in skin pigmentation as a variable to advance the equitability of pharmacological interventions. The article provides guidelines on the selection of New Approach Methods (NAMs) to foster inclusive study designs in preclinical drug development pipelines, leading to an improved level of translatability to the clinic.
Collapse
Affiliation(s)
- Sophie Zaaijer
- Cornell Tech, New York, NY, USA.
- University of California Riverside, Riverside, CA, USA.
| | - Simon C Groen
- University of California Riverside, Riverside, CA, USA.
| |
Collapse
|
21
|
Bagdad Y, Sisquellas M, Arthur M, Miteva MA. Machine Learning and Deep Learning Models for Predicting Noncovalent Inhibitors of AmpC β-Lactamase. ACS OMEGA 2024; 9:41334-41342. [PMID: 39398163 PMCID: PMC11465252 DOI: 10.1021/acsomega.4c03834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 10/15/2024]
Abstract
Continuous use of antibiotics leads to the ability of bacteria to adapt by developing complex antibiotic resistance (AR) mechanisms. The synthesis of β-lactamases is a widely observed AR mechanism. The class C β-lactamase (AmpC) causes significant resistance toward β-lactam antibiotics, and new treatments are urgently needed. Noncovalent inhibitors have been developed against a broad spectrum of β-lactamases. In this study, we developed robust and accurate models for predicting noncovalent inhibitors of AmpC using large compound data sets and machine/deep learning modeling. We created support vector machine (SVM), random forest (RF), and feed-forward neural network (FFNN) classification models. The cross-validation (CV) accuracies varied between 80 and 82%, as combined models reached an accuracy of 83%. We analyzed the physicochemical characteristics of the noncovalent inhibitors and predicted the binding modes for some of them. Such models are helpful for identifying new noncovalent inhibitors in order to establish novel solutions against the growing resistance to standard β-lactam inhibitors. The best RF, SVM, and FFNN models for predicting noncovalent inhibitors of AmpC β-lactamase are available in the GitHub repository, https://github.com/UPCmctr/ML-DL-AmpC-B-lactamase.
Collapse
Affiliation(s)
- Youcef Bagdad
- Université
Paris Cité, CNRS UMR 8038 CiTCoM, Inserm U1268 MCTR, 75006 Paris, France
| | - Marion Sisquellas
- Université
Paris Cité, CNRS UMR 8038 CiTCoM, Inserm U1268 MCTR, 75006 Paris, France
| | - Michel Arthur
- INSERM,
Sorbonne Université, Université Paris Cité, Centre
de Recherche des Cordeliers (CRC), 75006 Paris, France
| | - Maria A. Miteva
- Université
Paris Cité, CNRS UMR 8038 CiTCoM, Inserm U1268 MCTR, 75006 Paris, France
| |
Collapse
|
22
|
Ahmad S, Ahmad MFA, Alouffi S, Khan S, Khan M, Khan MWA, Prakash C, Ahmad N, Ansari IA. Aldose reductase inhibitory and antiglycation properties of phytoconstituents of Cichorium intybus: Potential therapeutic role in diabetic retinopathy. Int J Biol Macromol 2024; 277:133816. [PMID: 39002911 DOI: 10.1016/j.ijbiomac.2024.133816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 07/15/2024]
Abstract
Diabetic vascular complication including diabetic retinopathy is a major morbidity in Saudia Arabia. The polyol pathway aka aldose reductase (AR) pathway has gained significant association with diabetic retinopathy with regard to chronically enhanced glucose metabolism. Considerable research has been put forth to develop more effective therapeutic strategies to overcome the overwhelming challenges of vascular complications associated with diabetes. In this regard, constituents of Cichorium intybus can offer strong AR inhibitory potential because of their strong antidiabetic properties. Therefore, aim of this study was to investigate the AR inhibitory as well as antiglycation potential of C. intybus extract/compounds. The preliminary in vitro results showed that methanolic extract of C. intybus could significantly inhibit AR enzyme and advanced glycation end product formation. Eventually, based on previous studies and reviews, we selected one hundred fifteen C. intybus root constituents and screened them through Lipinski's rule of five and ADMET analysis. Later, after molecular docking analysis of eight compounds, five best were selected for molecular dynamics simulation to deduce their binding affinity with the AR enzyme. Finally, three out of five compounds were further tested in vitro for their AR inhibitory potential and antiglycation properties. Enzyme assay and kinetic studies showed that all the three tested compounds were having potent AR inhibitory properties, although to a lesser extent than ellagic acid and tolrestat. Similarly, kaempferol showed strong antiglycation property equivalent to ellagic acid, but greater than aminoguanidine. Intriguingly, significant reduction in sorbitol accumulation in RBCs by the tested compounds substantiated strong AR inhibition by these compounds. Moreover, decrease in sorbitol accumulation under high glucose environment also signifies the potential application of these compounds in diabetic retinopathy and other vascular complications. Thus, in sum, the in silico and in vitro studies combinedly showed that C. intybus root is a treasure for therapeutic compounds and can be explored further for drug development against diabetic retinopathy.
Collapse
Affiliation(s)
- Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail- 2440, Saudi Arabia.
| | | | - Sultan Alouffi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail- 2440, Saudi Arabia.
| | - Saif Khan
- Department of Basic Dental and Medical Sciences, College of Dentistry, University of Hail, Saudi Arabia.
| | - Mahvish Khan
- Department of Biology, College of Science, University of Hail- 2440, Saudi Arabia.
| | - Mohd Wajid Ali Khan
- Department of Chemistry, College of Science, University of Hail- 2440, Saudi Arabia.
| | - Chander Prakash
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India.
| | - Naved Ahmad
- Department of Computer Science and Information System, College of Applied Sciences, AlMaarefa University, P.O. Box 71666, Riyadh 13713, Saudi Arabia.
| | | |
Collapse
|
23
|
Kötter A, Allenspach S, Grebner C, Matter H, Hiss JA, Schneider G, Hessler G. Task-Similarity is a Crucial Factor for Few-Shot Meta-Learning of Structure-Activity Relationships. Chembiochem 2024; 25:e202400095. [PMID: 38682398 DOI: 10.1002/cbic.202400095] [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: 01/31/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
Machine learning models support computer-aided molecular design and compound optimization. However, the initial phases of drug discovery often face a scarcity of training data for these models. Meta-learning has emerged as a potentially promising strategy, harnessing the wealth of structure-activity data available for known targets to facilitate efficient few-shot model training for the specific target of interest. In this study, we assessed the effectiveness of two different meta-learning methods, namely model-agnostic meta-learning (MAML) and adaptive deep kernel fitting (ADKF), specifically in the regression setting. We investigated how factors such as dataset size and the similarity of training tasks impact predictability. The results indicate that ADKF significantly outperformed both MAML and a single-task baseline model on the inhibition data. However, the performance of ADKF varied across different test tasks. Our findings suggest that considerable enhancements in performance can be anticipated primarily when the task of interest is similar to the tasks incorporated in the meta-learning process.
Collapse
Affiliation(s)
- Alex Kötter
- R&D, Integrated Drug Discovery, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Stephan Allenspach
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Christoph Grebner
- R&D, Integrated Drug Discovery, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Hans Matter
- R&D, Integrated Drug Discovery, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Jan A Hiss
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Gisbert Schneider
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Gerhard Hessler
- R&D, Integrated Drug Discovery, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| |
Collapse
|
24
|
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
|
25
|
He G, Liu S, Liu Z, Wang C, Zhang K, Li H. Prototype-based contrastive substructure identification for molecular property prediction. Brief Bioinform 2024; 25:bbae565. [PMID: 39494969 PMCID: PMC11533112 DOI: 10.1093/bib/bbae565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/11/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Substructure-based representation learning has emerged as a powerful approach to featurize complex attributed graphs, with promising results in molecular property prediction (MPP). However, existing MPP methods mainly rely on manually defined rules to extract substructures. It remains an open challenge to adaptively identify meaningful substructures from numerous molecular graphs to accommodate MPP tasks. To this end, this paper proposes Prototype-based cOntrastive Substructure IdentificaTion (POSIT), a self-supervised framework to autonomously discover substructural prototypes across graphs so as to guide end-to-end molecular fragmentation. During pre-training, POSIT emphasizes two key aspects of substructure identification: firstly, it imposes a soft connectivity constraint to encourage the generation of topologically meaningful substructures; secondly, it aligns resultant substructures with derived prototypes through a prototype-substructure contrastive clustering objective, ensuring attribute-based similarity within clusters. In the fine-tuning stage, a cross-scale attention mechanism is designed to integrate substructure-level information to enhance molecular representations. The effectiveness of the POSIT framework is demonstrated by experimental results from diverse real-world datasets, covering both classification and regression tasks. Moreover, visualization analysis validates the consistency of chemical priors with identified substructures. The source code is publicly available at https://github.com/VRPharmer/POSIT.
Collapse
Affiliation(s)
- Gaoqi He
- School of Computer Science and Technology, East China Normal University, 200062 Shanghai, China
| | - Shun Liu
- School of Computer Science and Technology, East China Normal University, 200062 Shanghai, China
| | - Zhuoran Liu
- School of Computer Science and Technology, East China Normal University, 200062 Shanghai, China
| | - Changbo Wang
- School of Computer Science and Technology, East China Normal University, 200062 Shanghai, China
| | - Kai Zhang
- School of Computer Science and Technology, East China Normal University, 200062 Shanghai, China
| | - Honglin Li
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062 Shanghai, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science & Technology, 200237 Shanghai, China
| |
Collapse
|
26
|
Panjla A, Joshi S, Singh G, Bamford SE, Mechler A, Verma S. Applying Machine Learning for Antibiotic Development and Prediction of Microbial Resistance. Chem Asian J 2024; 19:e202400102. [PMID: 38948939 DOI: 10.1002/asia.202400102] [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: 01/30/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/02/2024]
Abstract
Antimicrobial resistance (AMR) poses a serious threat to human health worldwide. It is now more challenging than ever to introduce a potent antibiotic to the market considering rapid emergence of antimicrobial resistance, surpassing the rate of antibiotic drug discovery. Hence, new approaches need to be developed to accelerate the rate of drug discovery process and meet the demands for new antibiotics, while reducing the cost of their development. Machine learning holds immense promise of becoming a useful tool, especially since in the last two decades, exponential growth has occurred in computational power and biological big data analytics. Recent advancements in machine learning algorithms for drug discovery have provided significant clues for potential antibiotic classes. Apart from discovery of new scaffolds, the machine learning protocols will significantly impact prediction of AMR patterns and drug metabolism. In this review, we outline power of machine learning in antibiotic drug discovery, metabolic fate, and AMR prediction to support researchers engaged and interested in this field.
Collapse
Affiliation(s)
- Apurva Panjla
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Saurabh Joshi
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Geetanjali Singh
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Sarah E Bamford
- Department of Chemistry and Physics, La Trobe University, Bundoora, Victoria, 3086, Australia
| | - Adam Mechler
- Department of Chemistry and Physics, La Trobe University, Bundoora, Victoria, 3086, Australia
| | - Sandeep Verma
- Mehta Family Center for Engineering in Medicine, Center for Nanoscience, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| |
Collapse
|
27
|
Madushanka A, Laird E, Clark C, Kraka E. SmartCADD: AI-QM Empowered Drug Discovery Platform with Explainability. J Chem Inf Model 2024; 64:6799-6813. [PMID: 39177478 DOI: 10.1021/acs.jcim.4c00720] [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: 08/24/2024]
Abstract
Artificial intelligence (AI) has emerged as a pivotal force in enhancing productivity across various sectors, with its impact being profoundly felt within the pharmaceutical and biotechnology domains. Despite AI's rapid adoption, its integration into scientific research faces resistance due to myriad challenges: the opaqueness of AI models, the intricate nature of their implementation, and the issue of data scarcity. In response to these impediments, we introduce SmartCADD, an innovative, open-source virtual screening platform that combines deep learning, computer-aided drug design (CADD), and quantum mechanics methodologies within a user-friendly Python framework. SmartCADD is engineered to streamline the construction of comprehensive virtual screening workflows that incorporate a variety of formerly independent techniques─spanning ADMET property predictions, de novo 2D and 3D pharmacophore modeling, molecular docking, to the integration of explainable AI mechanisms. This manuscript highlights the foundational principles, key functionalities, and the unique integrative approach of SmartCADD. Furthermore, we demonstrate its efficacy through a case study focused on the identification of promising lead compounds for HIV inhibition. By democratizing access to advanced AI and quantum mechanics tools, SmartCADD stands as a catalyst for progress in pharmaceutical research and development, heralding a new era of innovation and efficiency.
Collapse
Affiliation(s)
- Ayesh Madushanka
- Department of Chemistry, Southern Methodist University, Dallas, Texas 75205, United States
| | - Eli Laird
- Department of Computer Science, Southern Methodist University, Dallas, Texas 75205, United States
| | - Corey Clark
- Department of Computer Science, Southern Methodist University, Dallas, Texas 75205, United States
| | - Elfi Kraka
- Department of Chemistry, Southern Methodist University, Dallas, Texas 75205, United States
| |
Collapse
|
28
|
Loeffler HH, Wan S, Klähn M, Bhati AP, Coveney PV. Optimal Molecular Design: Generative Active Learning Combining REINVENT with Precise Binding Free Energy Ranking Simulations. J Chem Theory Comput 2024; 20. [PMID: 39225482 PMCID: PMC11428133 DOI: 10.1021/acs.jctc.4c00576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/08/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Active learning (AL) is a specific instance of sequential experimental design and uses machine learning to intelligently choose the next data point or batch of molecular structures to be evaluated. In this sense, it closely mimics the iterative design-make-test-analysis cycle of laboratory experiments to find optimized compounds for a given design task. Here, we describe an AL protocol which combines generative molecular AI, using REINVENT, and physics-based absolute binding free energy molecular dynamics simulation, using ESMACS, to discover new ligands for two different target proteins, 3CLpro and TNKS2. We have deployed our generative active learning (GAL) protocol on Frontier, the world's only exa-scale machine. We show that the protocol can find higher-scoring molecules compared to the baseline, a surrogate ML docking model for 3CLpro and compounds with experimentally determined binding affinities for TNKS2. The ligands found are also chemically diverse and occupy a different chemical space than the baseline. We vary the batch sizes that are put forward for free energy assessment in each GAL cycle to assess the impact on their efficiency on the GAL protocol and recommend their optimal values in different scenarios. Overall, we demonstrate a powerful capability of the combination of physics-based and AI methods which yields effective chemical space sampling at an unprecedented scale and is of immediate and direct relevance to modern, data-driven drug discovery.
Collapse
Affiliation(s)
- Hannes H. Loeffler
- Molecular
AI, Discovery Sciences, R&D, AstraZeneca, Mölndal 431 83, Sweden
| | - Shunzhou Wan
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, U.K.
| | - Marco Klähn
- Molecular
AI, Discovery Sciences, R&D, AstraZeneca, Mölndal 431 83, Sweden
| | - Agastya P. Bhati
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, U.K.
| | - Peter V. Coveney
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, U.K.
- Advanced
Research Computing Centre, University College
London, London WC1H 0AJ, U.K.
- Institute
for Informatics, Faculty of Science, University
of Amsterdam, Amsterdam 1098XH, The Netherlands
| |
Collapse
|
29
|
Gangwal A, Ansari A, Ahmad I, Azad AK, Wan Sulaiman WMA. Current strategies to address data scarcity in artificial intelligence-based drug discovery: A comprehensive review. Comput Biol Med 2024; 179:108734. [PMID: 38964243 DOI: 10.1016/j.compbiomed.2024.108734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/01/2024] [Accepted: 06/08/2024] [Indexed: 07/06/2024]
Abstract
Artificial intelligence (AI) has played a vital role in computer-aided drug design (CADD). This development has been further accelerated with the increasing use of machine learning (ML), mainly deep learning (DL), and computing hardware and software advancements. As a result, initial doubts about the application of AI in drug discovery have been dispelled, leading to significant benefits in medicinal chemistry. At the same time, it is crucial to recognize that AI is still in its infancy and faces a few limitations that need to be addressed to harness its full potential in drug discovery. Some notable limitations are insufficient, unlabeled, and non-uniform data, the resemblance of some AI-generated molecules with existing molecules, unavailability of inadequate benchmarks, intellectual property rights (IPRs) related hurdles in data sharing, poor understanding of biology, focus on proxy data and ligands, lack of holistic methods to represent input (molecular structures) to prevent pre-processing of input molecules (feature engineering), etc. The major component in AI infrastructure is input data, as most of the successes of AI-driven efforts to improve drug discovery depend on the quality and quantity of data, used to train and test AI algorithms, besides a few other factors. Additionally, data-gulping DL approaches, without sufficient data, may collapse to live up to their promise. Current literature suggests a few methods, to certain extent, effectively handle low data for better output from the AI models in the context of drug discovery. These are transferring learning (TL), active learning (AL), single or one-shot learning (OSL), multi-task learning (MTL), data augmentation (DA), data synthesis (DS), etc. One different method, which enables sharing of proprietary data on a common platform (without compromising data privacy) to train ML model, is federated learning (FL). In this review, we compare and discuss these methods, their recent applications, and limitations while modeling small molecule data to get the improved output of AI methods in drug discovery. Article also sums up some other novel methods to handle inadequate data.
Collapse
Affiliation(s)
- Amit Gangwal
- Department of Natural Product Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India.
| | - Azim Ansari
- Computer Aided Drug Design Center, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Gondur, Dhule, 424002, Maharashtra, India.
| | - Abul Kalam Azad
- Faculty of Pharmacy, University College of MAIWP International, Batu Caves, 68100, Kuala Lumpur, Malaysia.
| | | |
Collapse
|
30
|
Kolesnikova TO, Demin KA, Costa FV, de Abreu MS, Kalueff AV. Zebrafish models for studying cognitive enhancers. Neurosci Biobehav Rev 2024; 164:105797. [PMID: 38971515 DOI: 10.1016/j.neubiorev.2024.105797] [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: 05/08/2024] [Revised: 06/16/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Cognitive decline is commonly seen both in normal aging and in neurodegenerative and neuropsychiatric diseases. Various experimental animal models represent a valuable tool to study brain cognitive processes and their deficits. Equally important is the search for novel drugs to treat cognitive deficits and improve cognitions. Complementing rodent and clinical findings, studies utilizing zebrafish (Danio rerio) are rapidly gaining popularity in translational cognitive research and neuroactive drug screening. Here, we discuss the value of zebrafish models and assays for screening nootropic (cognitive enhancer) drugs and the discovery of novel nootropics. We also discuss the existing challenges, and outline future directions of research in this field.
Collapse
Affiliation(s)
| | - Konstantin A Demin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Fabiano V Costa
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
| | - Murilo S de Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; West Caspian University, Baku, Azerbaijan.
| | - Allan V Kalueff
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Suzhou Key Laboratory on Neurobiology and Cell Signaling, Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China.
| |
Collapse
|
31
|
Barrera Patiño CP, Soares JM, Blanco KC, Bagnato VS. Machine Learning in FTIR Spectrum for the Identification of Antibiotic Resistance: A Demonstration with Different Species of Microorganisms. Antibiotics (Basel) 2024; 13:821. [PMID: 39334995 PMCID: PMC11428736 DOI: 10.3390/antibiotics13090821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/22/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Recent studies introduced the importance of using machine learning algorithms in research focused on the identification of antibiotic resistance. In this study, we highlight the importance of building solid machine learning foundations to differentiate antimicrobial resistance among microorganisms. Using advanced machine learning algorithms, we established a methodology capable of analyzing the FTIR structural profile of the samples of Streptococcus pyogenes and Streptococcus mutans (Gram-positive), as well as Escherichia coli and Klebsiella pneumoniae (Gram-negative), demonstrating cross-sectional applicability in this focus on different microorganisms. The analysis focuses on specific biomolecules-Carbohydrates, Fatty Acids, and Proteins-in FTIR spectra, providing a multidimensional database that transcends microbial variability. The results highlight the ability of the method to consistently identify resistance patterns, regardless of the Gram classification of the bacteria and the species involved, reinforcing the premise that the structural characteristics identified are universal among the microorganisms tested. By validating this approach in four distinct species, our study proves the versatility and precision of the methodology used, in addition to bringing support to the development of an innovative protocol for the rapid and safe identification of antimicrobial resistance. This advance is crucial for optimizing treatment strategies and avoiding the spread of resistance. This emphasizes the relevance of specialized machine learning bases in effectively differentiating between resistance profiles in Gram-negative and Gram-positive bacteria to be implemented in the identification of antibiotic resistance. The obtained result has a high potential to be applied to clinical procedures.
Collapse
Affiliation(s)
- Claudia Patricia Barrera Patiño
- São Carlos Institute of Physics, University of São Paulo, Avenida Trabalhador São-Carlense No. 400, Parque Arnold Schimidt, São Carlos CEP 13566-590, SP, Brazil
| | - Jennifer Machado Soares
- São Carlos Institute of Physics, University of São Paulo, Avenida Trabalhador São-Carlense No. 400, Parque Arnold Schimidt, São Carlos CEP 13566-590, SP, Brazil
| | - Kate Cristina Blanco
- São Carlos Institute of Physics, University of São Paulo, Avenida Trabalhador São-Carlense No. 400, Parque Arnold Schimidt, São Carlos CEP 13566-590, SP, Brazil
| | - Vanderlei Salvador Bagnato
- São Carlos Institute of Physics, University of São Paulo, Avenida Trabalhador São-Carlense No. 400, Parque Arnold Schimidt, São Carlos CEP 13566-590, SP, Brazil
- Biomedical Engineering, Texas A&M University, 400 Bizzell St., College Station, TX 77843, USA
| |
Collapse
|
32
|
Zheng Y, Ma Y, Xiong Q, Zhu K, Weng N, Zhu Q. The role of artificial intelligence in the development of anticancer therapeutics from natural polyphenols: Current advances and future prospects. Pharmacol Res 2024; 208:107381. [PMID: 39218422 DOI: 10.1016/j.phrs.2024.107381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/06/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Natural polyphenols, abundant in the human diet, are derived from a wide variety of sources. Numerous preclinical studies have demonstrated their significant anticancer properties against various malignancies, making them valuable resources for drug development. However, traditional experimental methods for developing anticancer therapies from natural polyphenols are time-consuming and labor-intensive. Recently, artificial intelligence has shown promising advancements in drug discovery. Integrating AI technologies into the development process for natural polyphenols can substantially reduce development time and enhance efficiency. In this study, we review the crucial roles of natural polyphenols in anticancer treatment and explore the potential of AI technologies to aid in drug development. Specifically, we discuss the application of AI in key stages such as drug structure prediction, virtual drug screening, prediction of biological activity, and drug-target protein interaction, highlighting the potential to revolutionize the development of natural polyphenol-based anticancer therapies.
Collapse
Affiliation(s)
- Ying Zheng
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Yifei Ma
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Qunli Xiong
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Kai Zhu
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian 350011, PR China
| | - Ningna Weng
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian 350011, PR China
| | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China.
| |
Collapse
|
33
|
Garnsey MR, Robinson MC, Nguyen LT, Cardin R, Tillotson J, Mashalidis E, Yu A, Aschenbrenner L, Balesano A, Behzadi A, Boras B, Chang JS, Eng H, Ephron A, Foley T, Ford KK, Frick JM, Gibson S, Hao L, Hurst B, Kalgutkar AS, Korczynska M, Lengyel-Zhand Z, Gao L, Meredith HR, Patel NC, Polivkova J, Rai D, Rose CR, Rothan H, Sakata SK, Vargo TR, Qi W, Wu H, Liu Y, Yurgelonis I, Zhang J, Zhu Y, Zhang L, Lee AA. Discovery of SARS-CoV-2 papain-like protease (PL pro) inhibitors with efficacy in a murine infection model. SCIENCE ADVANCES 2024; 10:eado4288. [PMID: 39213347 PMCID: PMC11364104 DOI: 10.1126/sciadv.ado4288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Vaccines and first-generation antiviral therapeutics have provided important protection against COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, there remains a need for additional therapeutic options that provide enhanced efficacy and protection against potential viral resistance. The SARS-CoV-2 papain-like protease (PLpro) is one of the two essential cysteine proteases involved in viral replication. While inhibitors of the SARS-CoV-2 main protease have demonstrated clinical efficacy, known PLpro inhibitors have, to date, lacked the inhibitory potency and requisite pharmacokinetics to demonstrate that targeting PLpro translates to in vivo efficacy in a preclinical setting. Here, we report the machine learning-driven discovery of potent, selective, and orally available SARS-CoV-2 PLpro inhibitors, with lead compound PF-07957472 (4) providing robust efficacy in a mouse-adapted model of COVID-19 infection.
Collapse
Affiliation(s)
| | | | | | - Rhonda Cardin
- Pfizer Global Research and Development, Pearl River, NY 10965, USA
| | - Joseph Tillotson
- Pfizer Global Research and Development, Cambridge, MA 02139, USA
| | | | - Aijia Yu
- WuXi, WuXi AppTec (Shanghai) Co. Ltd. Shanghai 200131, China
| | | | - Amanda Balesano
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Amin Behzadi
- Pfizer Global Research and Development, Pearl River, NY 10965, USA
| | - Britton Boras
- Pfizer Global Research and Development, La Jolla, CA 92121, USA
| | - Jeanne S. Chang
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Heather Eng
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Andrew Ephron
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Tim Foley
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Kristen K. Ford
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - James M. Frick
- PostEra, 1 Broadway, 14th floor, Cambridge, MA 02142, USA
| | - Scott Gibson
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences,Utah State University, Logan, UT 84322, USA
| | - Li Hao
- Pfizer Global Research and Development, Pearl River, NY 10965, USA
| | - Brett Hurst
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences,Utah State University, Logan, UT 84322, USA
| | | | | | | | - Liping Gao
- WuXi, WuXi AppTec (Shanghai) Co. Ltd. Shanghai 200131, China
| | | | - Nandini C. Patel
- Pfizer Global Research and Development, Cambridge, MA 02139, USA
| | - Jana Polivkova
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Devendra Rai
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Colin R. Rose
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Hussin Rothan
- Pfizer Global Research and Development, Pearl River, NY 10965, USA
| | | | | | - Wenying Qi
- WuXi, WuXi AppTec (Shanghai) Co. Ltd. Shanghai 200131, China
| | - Huixian Wu
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Yiping Liu
- WuXi, WuXi AppTec (Shanghai) Co. Ltd. Shanghai 200131, China
| | - Irina Yurgelonis
- Pfizer Global Research and Development, Pearl River, NY 10965, USA
| | - Jinzhi Zhang
- Pfizer Global Research and Development, Shanghai 201210, China
| | - Yuao Zhu
- Pfizer Global Research and Development, Pearl River, NY 10965, USA
| | - Lei Zhang
- Pfizer Global Research and Development, Cambridge, MA 02139, USA
| | - Alpha A. Lee
- PostEra, 1 Broadway, 14th floor, Cambridge, MA 02142, USA
| |
Collapse
|
34
|
Wang B, Guo J, Chen B, Jiao Y, Wan Y, Wu J, Wang Y. Combination of ligand‑based and structure‑based virtual screening for the discovery of novel Janus kinase 2 inhibitors against philadelphia-negative myeloproliferative neoplasms. Mol Divers 2024:10.1007/s11030-024-10938-1. [PMID: 39210217 DOI: 10.1007/s11030-024-10938-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 07/10/2024] [Indexed: 09/04/2024]
Abstract
The activating V617F mutation in Janus kinase 2 (JAK2) has been shown to be the major cause for classic Philadelphia-negative myeloproliferative neoplasms (MPNs). Thus, the development of pharmacologic JAK2 inhibitors is an essential move in combating MPNs. In this study, screening methods examining both ligands and their structures were developed to discover novel JAK2 inhibitors from the ChemDiv database with virtual screening identifying 886 candidate inhibitors. Next, these compounds were further filtered using ADMET, drug likeliness, and PAINS filtering, which reduced the compound number even further. This consolidated list of candidate compounds (n = 49) was then evaluated biologically at molecular level and the highest performing inhibitor with a novel scaffold was selected for further examination. This candidate inhibitor, CD4, was then subjected to molecular dynamics studies, with complex stability, root-mean-square deviation, radius of gyration, binding free energy, and binding properties all examined. The result suggested that CD4 interacts with JAK2 and that the CD4-JAK2 complex is stable. This study was able to identify a candidate inhibitor that warrants further examination and optimization and may potentially serve as a future MPN treatment.
Collapse
Affiliation(s)
- Binyou Wang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
- Zigong Mental Health Center, Zigong Affiliated Hospital of Southwest Medical University, Zigong, 643000, China
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Jianmin Guo
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Bo Chen
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Yan Jiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Jianming Wu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - Yiwei Wang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
35
|
Pais DAM, Mayer JPA, Felderer K, Batalha MB, Eichner T, Santos ST, Kumar R, Silva SD, Kaufmann H. Holistic in silico developability assessment of novel classes of small proteins using publicly available sequence-based predictors. J Comput Aided Mol Des 2024; 38:30. [PMID: 39164492 DOI: 10.1007/s10822-024-00569-x] [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: 01/16/2024] [Accepted: 07/26/2024] [Indexed: 08/22/2024]
Abstract
The development of novel therapeutic proteins is a lengthy and costly process, with an average attrition rate of 91% (Thomas et al. Clinical Development Success Rates and Contributing Factors 2011-2020, 2021). To increase the probability of success and ensure robust drug supply beyond approval, it is essential to assess the developability profile of new potential drug candidates as early and broadly as possible in development (Jain et al. MAbs, 2023. https://doi.org/10.1016/j.copbio.2011.06.002 ). Predicting these properties in silico is expected to be the next leap in innovation as it would enable significantly reduced development timelines combined with broader screens at lower costs. However, developing predictive algorithms typically requires substantial datasets generated under very defined conditions, a limiting factor especially for new classes of therapeutic proteins that hold immense clinical promise. Here we describe a strategy for assessing the developability of a novel class of small therapeutic Anticalin® proteins using machine learning in conjunction with a knowledge-driven approach. The knowledge-driven approach considers developability attributes such as aggregation propensity, charge variants, immunogenicity, specificity, thermal stability, hydrophobicity, and potential post-translational modifications, to calculate a holistic developability score. Based on sequence-derived descriptors as input parameters we established novel statistical models designed to predict the developability scores for Anticalin proteins. The best models yielded low root mean square errors across the entire dataset and were further validated by removing input data from individual screening campaigns and predicting developability scores for those drug candidates. The adoption of the described workflow will enable significantly streamlined preclinical development of Anticalin drug candidates and could potentially be applied to other therapeutic protein scaffolds.
Collapse
Affiliation(s)
- Daniel A M Pais
- Valgenesis Portugal, Lda, R. Castilho 50 4th Floor, 1250-071, Lisbon, Portugal
| | - Jan-Peter A Mayer
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany
| | - Karin Felderer
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany
| | - Maria B Batalha
- Valgenesis Portugal, Lda, R. Castilho 50 4th Floor, 1250-071, Lisbon, Portugal
| | - Timo Eichner
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany
| | - Sofia T Santos
- Valgenesis Portugal, Lda, R. Castilho 50 4th Floor, 1250-071, Lisbon, Portugal
| | - Raman Kumar
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany
| | - Sandra D Silva
- Valgenesis Portugal, Lda, R. Castilho 50 4th Floor, 1250-071, Lisbon, Portugal
| | - Hitto Kaufmann
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany.
| |
Collapse
|
36
|
Rezić I, Somogyi Škoc M. Computational Methodologies in Synthesis, Preparation and Application of Antimicrobial Polymers, Biomolecules, and Nanocomposites. Polymers (Basel) 2024; 16:2320. [PMID: 39204538 PMCID: PMC11359845 DOI: 10.3390/polym16162320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/05/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
The design and optimization of antimicrobial materials (polymers, biomolecules, or nanocomposites) can be significantly advanced by computational methodologies like molecular dynamics (MD), which provide insights into the interactions and stability of the antimicrobial agents within the polymer matrix, and machine learning (ML) or design of experiment (DOE), which predicts and optimizes antimicrobial efficacy and material properties. These innovations not only enhance the efficiency of developing antimicrobial polymers but also enable the creation of materials with tailored properties to meet specific application needs, ensuring safety and longevity in their usage. Therefore, this paper will present the computational methodologies employed in the synthesis and application of antimicrobial polymers, biomolecules, and nanocomposites. By leveraging advanced computational techniques such as MD, ML, or DOE, significant advancements in the design and optimization of antimicrobial materials are achieved. A comprehensive review on recent progress, together with highlights of the most relevant methodologies' contributions to state-of-the-art materials science will be discussed, as well as future directions in the field will be foreseen. Finally, future possibilities and opportunities will be derived from the current state-of-the-art methodologies, providing perspectives on the potential evolution of polymer science and engineering of novel materials.
Collapse
Affiliation(s)
- Iva Rezić
- Department of Applied Chemistry, Faculty of Textile Technology, University of Zagreb, 10000 Zagreb, Croatia
| | - Maja Somogyi Škoc
- Department of Materials Testing, Faculty of Textile Technology, University of Zagreb, 10000 Zagreb, Croatia;
| |
Collapse
|
37
|
Chenab KK, Malektaj H, Nadinlooie AAR, Mohammadi S, Zamani-Meymian MR. Intertumoral and intratumoral barriers as approaches for drug delivery and theranostics to solid tumors using stimuli-responsive materials. Mikrochim Acta 2024; 191:541. [PMID: 39150483 DOI: 10.1007/s00604-024-06583-y] [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: 05/01/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
The solid tumors provide a series of biological barriers in cellular microenvironment for designing drug delivery methods based on advanced stimuli-responsive materials. These intertumoral and intratumoral barriers consist of perforated endotheliums, tumor cell crowding, vascularity, lymphatic drainage blocking effect, extracellular matrix (ECM) proteins, hypoxia, and acidosis. Triggering opportunities have been drawn for solid tumor therapies based on single and dual stimuli-responsive drug delivery systems (DDSs) that not only improved drug targeting in deeper sites of the tumor microenvironments, but also facilitated the antitumor drug release efficiency. Single and dual stimuli-responsive materials which are known for their lowest side effects can be categorized in 17 main groups which involve to internal and external stimuli anticancer drug carriers in proportion to microenvironments of targeted solid tumors. Development of such drug carriers can circumvent barriers in clinical trial studies based on their superior capabilities in penetrating into more inaccessible sites of the tumor tissues. In recent designs, key characteristics of these DDSs such as fast response to intracellular and extracellular factors, effective cytotoxicity with minimum side effect, efficient permeability, and rate and location of drug release have been discussed as core concerns of designing paradigms of these materials.
Collapse
Affiliation(s)
- Karim Khanmohammadi Chenab
- Department of Chemistry, Iran University of Science and Technology, Tehran, P.O. Box 16846-13114, Iran
- Department of Physics, Iran University of Science and Technology, Tehran, P.O. Box 16846-13114, Iran
| | - Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, 9220, Aalborg, Denmark
| | | | | | | |
Collapse
|
38
|
Tandon A, Santura A, Waldmann H, Pahl A, Czodrowski P. Identification of lysosomotropism using explainable machine learning and morphological profiling cell painting data. RSC Med Chem 2024; 15:2677-2691. [PMID: 39149097 PMCID: PMC11324048 DOI: 10.1039/d4md00107a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/09/2024] [Indexed: 08/17/2024] Open
Abstract
Lysosomotropism is a phenomenon of diverse pharmaceutical interests because it is a property of compounds with diverse chemical structures and primary targets. While it is primarily reported to be caused by compounds having suitable lipophilicity and basicity values, not all compounds that fulfill such criteria are in fact lysosomotropic. Here, we use morphological profiling by means of the cell painting assay (CPA) as a reliable surrogate to identify lysosomotropism. We noticed that only 35% of the compound subset with matching physicochemical properties show the lysosomotropic phenotype. Based on a matched molecular pair analysis (MMPA), no key substructures driving lysosomotropism could be identified. However, using explainable machine learning (XML), we were able to highlight that higher lipophilicity, basicity, molecular weight, and lower topological polar surface area are among the important properties that induce lysosomotropism in the compounds of this subset.
Collapse
Affiliation(s)
- Aishvarya Tandon
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology Otto-Hahn-Str. 11 Dortmund Germany
| | - Anna Santura
- Department of Chemistry, Johannes Gutenberg University Mainz Mainz Germany
| | - Herbert Waldmann
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology Otto-Hahn-Str. 11 Dortmund Germany
| | - Axel Pahl
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology Otto-Hahn-Str. 11 Dortmund Germany
| | - Paul Czodrowski
- Department of Chemistry, Johannes Gutenberg University Mainz Mainz Germany
| |
Collapse
|
39
|
Oualikene-Gonin W, Jaulent MC, Thierry JP, Oliveira-Martins S, Belgodère L, Maison P, Ankri J. Artificial intelligence integration in the drug lifecycle and in regulatory science: policy implications, challenges and opportunities. Front Pharmacol 2024; 15:1437167. [PMID: 39156111 PMCID: PMC11327028 DOI: 10.3389/fphar.2024.1437167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/18/2024] [Indexed: 08/20/2024] Open
Abstract
Artificial intelligence tools promise transformative impacts in drug development. Regulatory agencies face challenges in integrating AI while ensuring reliability and safety in clinical trial approvals, drug marketing authorizations, and post-market surveillance. Incorporating these technologies into the existing regulatory framework and agency practices poses notable challenges, particularly in evaluating the data and models employed for these purposes. Rapid adaptation of regulations and internal processes is essential for agencies to keep pace with innovation, though achieving this requires collective stakeholder collaboration. This article thus delves into the need for adaptations of regulations throughout the drug development lifecycle, as well as the utilization of AI within internal processes of medicine agencies.
Collapse
Affiliation(s)
- Wahiba Oualikene-Gonin
- Agence Nationale de Sécurité des Médicaments et des Produits de Santé (ANSM) Saint-Denis, Saint-Denis, France
| | - Marie-Christine Jaulent
- INSERM, Laboratoire d'Informatique Médicale et d'Ingénierie des Connaissances en e-Santé, LIMICS, Sorbonne Université, Paris, France
| | | | - Sofia Oliveira-Martins
- Faculty of Pharmacy of Lisbon University, Lisbon, Portugal
- CHRC – Comprehensive Health Research Center, Evora, Portugal
| | - Laetitia Belgodère
- Agence Nationale de Sécurité des Médicaments et des Produits de Santé (ANSM) Saint-Denis, Saint-Denis, France
| | - Patrick Maison
- Agence Nationale de Sécurité des Médicaments et des Produits de Santé (ANSM) Saint-Denis, Saint-Denis, France
- EA 7379, Faculté de Santé, Université Paris-Est Créteil, Créteil, France
- CHI Créteil, Créteil, France
| | - Joël Ankri
- Université de Versailles St Quentin-Paris Saclay, Inserm U1018, Guyancourt, France
| | | |
Collapse
|
40
|
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
|
41
|
Lv Q, Chen G, Yang Z, Zhong W, Chen CYC. Meta Learning With Graph Attention Networks for Low-Data Drug Discovery. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2024; 35:11218-11230. [PMID: 37028032 DOI: 10.1109/tnnls.2023.3250324] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Finding candidate molecules with favorable pharmacological activity, low toxicity, and proper pharmacokinetic properties is an important task in drug discovery. Deep neural networks have made impressive progress in accelerating and improving drug discovery. However, these techniques rely on a large amount of label data to form accurate predictions of molecular properties. At each stage of the drug discovery pipeline, usually, only a few biological data of candidate molecules and derivatives are available, indicating that the application of deep neural networks for low-data drug discovery is still a formidable challenge. Here, we propose a meta learning architecture with graph attention network, Meta-GAT, to predict molecular properties in low-data drug discovery. The GAT captures the local effects of atomic groups at the atom level through the triple attentional mechanism and implicitly captures the interactions between different atomic groups at the molecular level. GAT is used to perceive molecular chemical environment and connectivity, thereby effectively reducing sample complexity. Meta-GAT further develops a meta learning strategy based on bilevel optimization, which transfers meta knowledge from other attribute prediction tasks to low-data target tasks. In summary, our work demonstrates how meta learning can reduce the amount of data required to make meaningful predictions of molecules in low-data scenarios. Meta learning is likely to become the new learning paradigm in low-data drug discovery. The source code is publicly available at: https://github.com/lol88/Meta-GAT.
Collapse
|
42
|
Pennington LD, Hesse MJ, Koester DC, McAtee RC, Qunies AM, Hu DX. Property-Based Drug Design Merits a Nobel Prize. J Med Chem 2024; 67:11452-11458. [PMID: 38940466 DOI: 10.1021/acs.jmedchem.4c01345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Affiliation(s)
| | | | | | - Rory C McAtee
- Drug Hunter, Happy Valley, Oregon 97086, United States
| | | | - Dennis X Hu
- Drug Hunter, Happy Valley, Oregon 97086, United States
| |
Collapse
|
43
|
Atz K, Nippa DF, Müller AT, Jost V, Anelli A, Reutlinger M, Kramer C, Martin RE, Grether U, Schneider G, Wuitschik G. Geometric deep learning-guided Suzuki reaction conditions assessment for applications in medicinal chemistry. RSC Med Chem 2024; 15:2310-2321. [PMID: 39026644 PMCID: PMC11253849 DOI: 10.1039/d4md00196f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/25/2024] [Indexed: 07/20/2024] Open
Abstract
Suzuki cross-coupling reactions are considered a valuable tool for constructing carbon-carbon bonds in small molecule drug discovery. However, the synthesis of chemical matter often represents a time-consuming and labour-intensive bottleneck. We demonstrate how machine learning methods trained on high-throughput experimentation (HTE) data can be leveraged to enable fast reaction condition selection for novel coupling partners. We show that the trained models support chemists in determining suitable catalyst-solvent-base combinations for individual transformations including an evaluation of the need for HTE screening. We introduce an algorithm for designing 96-well plates optimized towards reaction yields and discuss the model performance of zero- and few-shot machine learning. The best-performing machine learning model achieved a three-category classification accuracy of 76.3% (±0.2%) and an F 1-score for a binary classification of 79.1% (±0.9%). Validation on eight reactions revealed a receiver operating characteristic (ROC) curve (AUC) value of 0.82 (±0.07) for few-shot machine learning. On the other hand, zero-shot machine learning models achieved a mean ROC-AUC value of 0.63 (±0.16). This study positively advocates the application of few-shot machine learning-guided reaction condition selection for HTE campaigns in medicinal chemistry and highlights practical applications as well as challenges associated with zero-shot machine learning.
Collapse
Affiliation(s)
- Kenneth Atz
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Grenzacherstrasse 124 4070 Basel Switzerland
| | - David F Nippa
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Grenzacherstrasse 124 4070 Basel Switzerland
| | - Alex T Müller
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Grenzacherstrasse 124 4070 Basel Switzerland
| | - Vera Jost
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Grenzacherstrasse 124 4070 Basel Switzerland
| | - Andrea Anelli
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Grenzacherstrasse 124 4070 Basel Switzerland
| | - Michael Reutlinger
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Grenzacherstrasse 124 4070 Basel Switzerland
| | - Christian Kramer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Grenzacherstrasse 124 4070 Basel Switzerland
| | - Rainer E Martin
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Grenzacherstrasse 124 4070 Basel Switzerland
| | - Uwe Grether
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Grenzacherstrasse 124 4070 Basel Switzerland
| | - Gisbert Schneider
- Department of Chemistry and Applied Biosciences, ETH Zurich Vladimir-Prelog-Weg 4 8093 Zurich Switzerland
| | - Georg Wuitschik
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Grenzacherstrasse 124 4070 Basel Switzerland
| |
Collapse
|
44
|
Abbas MKG, Rassam A, Karamshahi F, Abunora R, Abouseada M. The Role of AI in Drug Discovery. Chembiochem 2024; 25:e202300816. [PMID: 38735845 DOI: 10.1002/cbic.202300816] [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: 12/03/2023] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024]
Abstract
The emergence of Artificial Intelligence (AI) in drug discovery marks a pivotal shift in pharmaceutical research, blending sophisticated computational techniques with conventional scientific exploration to break through enduring obstacles. This review paper elucidates the multifaceted applications of AI across various stages of drug development, highlighting significant advancements and methodologies. It delves into AI's instrumental role in drug design, polypharmacology, chemical synthesis, drug repurposing, and the prediction of drug properties such as toxicity, bioactivity, and physicochemical characteristics. Despite AI's promising advancements, the paper also addresses the challenges and limitations encountered in the field, including data quality, generalizability, computational demands, and ethical considerations. By offering a comprehensive overview of AI's role in drug discovery, this paper underscores the technology's potential to significantly enhance drug development, while also acknowledging the hurdles that must be overcome to fully realize its benefits.
Collapse
Affiliation(s)
- M K G Abbas
- Center for Advanced Materials, Qatar University, P.O. Box, 2713, Doha, Qatar
| | - Abrar Rassam
- Secondary Education, Educational Sciences, Qatar University, P.O. Box, 2713, Doha, Qatar
| | - Fatima Karamshahi
- Department of Chemistry and Earth Sciences, Qatar University, P.O. Box, 2713, Doha, Qatar
| | - Rehab Abunora
- Faculty of Medicine, General Medicine and Surgery, Helwan University, Cairo, Egypt
| | - Maha Abouseada
- Department of Chemistry and Earth Sciences, Qatar University, P.O. Box, 2713, Doha, Qatar
| |
Collapse
|
45
|
Bergasa-Caceres F, Rabitz HA. A Perspective on Interdicting in Protein Misfolding for Therapeutic Drug Design: Modulating the Formation of Nonlocal Contacts in α-Synuclein as a Strategy against Parkinson's Disease. J Phys Chem B 2024; 128:6439-6448. [PMID: 38940731 PMCID: PMC11247489 DOI: 10.1021/acs.jpcb.3c07519] [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: 11/14/2023] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/29/2024]
Abstract
In recent work we proposed that interdiction in the earliest contact-formation events along the folding pathway of key viral proteins could provide a novel avenue for therapeutic drug design. In this Perspective we explore the potential applicability of the protein folding interdiction strategy in the realm of neurodegenerative diseases with a specific focus on synucleinopathies. In order to fulfill this goal we review the interdiction proposal and its practical challenges, and we present new results concerning design strategies for possible peptide drugs that could be useful in preventing α-synuclein aggregation.
Collapse
Affiliation(s)
| | - Herschel A. Rabitz
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
46
|
An Y, Lim J, Glavatskikh M, Wang X, Norris-Drouin J, Hardy PB, Leisner TM, Pearce KH, Kireev D. In silico fragment-based discovery of CIB1-directed anti-tumor agents by FRASE-bot. Nat Commun 2024; 15:5564. [PMID: 38956119 PMCID: PMC11219766 DOI: 10.1038/s41467-024-49892-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 06/19/2024] [Indexed: 07/04/2024] Open
Abstract
Chemical probes are an indispensable tool for translating biological discoveries into new therapies, though are increasingly difficult to identify since novel therapeutic targets are often hard-to-drug proteins. We introduce FRASE-based hit-finding robot (FRASE-bot), to expedite drug discovery for unconventional therapeutic targets. FRASE-bot mines available 3D structures of ligand-protein complexes to create a database of FRAgments in Structural Environments (FRASE). The FRASE database can be screened to identify structural environments similar to those in the target protein and seed the target structure with relevant ligand fragments. A neural network model is used to retain fragments with the highest likelihood of being native binders. The seeded fragments then inform ultra-large-scale virtual screening of commercially available compounds. We apply FRASE-bot to identify ligands for Calcium and Integrin Binding protein 1 (CIB1), a promising drug target implicated in triple negative breast cancer. FRASE-based virtual screening identifies a small-molecule CIB1 ligand (with binding confirmed in a TR-FRET assay) showing specific cell-killing activity in CIB1-dependent cancer cells, but not in CIB1-depletion-insensitive cells.
Collapse
Affiliation(s)
- Yi An
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27513, USA
| | - Jiwoong Lim
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27513, USA
| | - Marta Glavatskikh
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27513, USA
| | - Xiaowen Wang
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27513, USA
- Chemistry department, University of Missouri, Columbia, Columbia, MO, 65211, USA
| | - Jacqueline Norris-Drouin
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27513, USA
| | - P Brian Hardy
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27513, USA
| | - Tina M Leisner
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27513, USA
| | - Kenneth H Pearce
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27513, USA.
| | - Dmitri Kireev
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27513, USA.
- Chemistry department, University of Missouri, Columbia, Columbia, MO, 65211, USA.
| |
Collapse
|
47
|
Zhou Y, Yao Z, Lin Y, Zhang H. From Tyrosine Kinases to Tyrosine Phosphatases: New Therapeutic Targets in Cancers and Beyond. Pharmaceutics 2024; 16:888. [PMID: 39065585 PMCID: PMC11279542 DOI: 10.3390/pharmaceutics16070888] [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: 04/25/2024] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs) regulate the level of tyrosine phosphorylation in proteins. PTKs are key enzymes that catalyze the transfer of an ATP phosphoric acid to a tyrosine residue on target protein substrates. Protein tyrosine phosphatases (PTPs) are responsible for the dephosphorylation of tyrosine residues and play a role in countering PTK overactivity. As widespread oncogenes, PTKs were once considered to be promising targets for therapy. However, tyrosine kinase inhibitors (TKIs) now face a number of challenges, including drug resistance and toxic side effects. Treatment strategies now need to be developed from a new perspective. In this review, we assess the current state of TKIs and highlight the role of PTPs in cancer and other diseases. With the advances of allosteric inhibition and the development of multiple alternative proprietary drug strategies, the reputation of PTPs as "undruggable" targets has been overturned, and they are now considered viable therapeutic targets. We also discuss the strategies and prospects of PTP-targeted therapy, as well as its future development.
Collapse
Affiliation(s)
- Yu Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou 510632, China; (Y.Z.); (Z.Y.); (Y.L.)
| | - Zhimeng Yao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou 510632, China; (Y.Z.); (Z.Y.); (Y.L.)
- Department of Urology Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510660, China
| | - Yusheng Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou 510632, China; (Y.Z.); (Z.Y.); (Y.L.)
- Department of Thoracic Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510660, China
| | - Hao Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou 510632, China; (Y.Z.); (Z.Y.); (Y.L.)
- Department of Pathology, Gongli Hospital of Shanghai Pudong New Area, Shanghai 200135, China
- Zhuhai Institute of Jinan University, Zhuhai 511436, China
| |
Collapse
|
48
|
Hao Y, Wang H, Liu X, Gai W, Hu S, Liu W, Miao Z, Gan Y, Yu X, Shi R, Tan Y, Kang T, Hai A, Zhao Y, Fu Y, Tang Y, Ye L, Liu J, Liang X, Ke B. Deep simulated annealing for the discovery of novel dental anesthetics with local anesthesia and anti-inflammatory properties. Acta Pharm Sin B 2024; 14:3086-3109. [PMID: 39027234 PMCID: PMC11252475 DOI: 10.1016/j.apsb.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/04/2024] [Accepted: 01/22/2024] [Indexed: 07/20/2024] Open
Abstract
Multifunctional therapeutics have emerged as a solution to the constraints imposed by drugs with singular or insufficient therapeutic effects. The primary challenge is to integrate diverse pharmacophores within a single-molecule framework. To address this, we introduced DeepSA, a novel edit-based generative framework that utilizes deep simulated annealing for the modification of articaine, a well-known local anesthetic. DeepSA integrates deep neural networks into metaheuristics, effectively constraining molecular space during compound generation. This framework employs a sophisticated objective function that accounts for scaffold preservation, anti-inflammatory properties, and covalent constraints. Through a sequence of local editing to navigate the molecular space, DeepSA successfully identified AT-17, a derivative exhibiting potent analgesic properties and significant anti-inflammatory activity in various animal models. Mechanistic insights into AT-17 revealed its dual mode of action: selective inhibition of NaV1.7 and 1.8 channels, contributing to its prolonged local anesthetic effects, and suppression of inflammatory mediators via modulation of the NLRP3 inflammasome pathway. These findings not only highlight the efficacy of AT-17 as a multifunctional drug candidate but also highlight the potential of DeepSA in facilitating AI-enhanced drug discovery, particularly within stringent chemical constraints.
Collapse
Affiliation(s)
- Yihang Hao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Haofan Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xianggen Liu
- College of Computer Science, Sichuan University, Chengdu 610065, China
| | - Wenrui Gai
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shilong Hu
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wencheng Liu
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhuang Miao
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Gan
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xianghua Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Rongjia Shi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yongzhen Tan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ting Kang
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ao Hai
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Zhao
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yihang Fu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yaling Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jin Liu
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinhua Liang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Bowen Ke
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
49
|
Iyer RR, Applegate CC, Arogundade OH, Bangru S, Berg IC, Emon B, Porras-Gomez M, Hsieh PH, Jeong Y, Kim Y, Knox HJ, Moghaddam AO, Renteria CA, Richard C, Santaliz-Casiano A, Sengupta S, Wang J, Zambuto SG, Zeballos MA, Pool M, Bhargava R, Gaskins HR. Inspiring a convergent engineering approach to measure and model the tissue microenvironment. Heliyon 2024; 10:e32546. [PMID: 38975228 PMCID: PMC11226808 DOI: 10.1016/j.heliyon.2024.e32546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024] Open
Abstract
Understanding the molecular and physical complexity of the tissue microenvironment (TiME) in the context of its spatiotemporal organization has remained an enduring challenge. Recent advances in engineering and data science are now promising the ability to study the structure, functions, and dynamics of the TiME in unprecedented detail; however, many advances still occur in silos that rarely integrate information to study the TiME in its full detail. This review provides an integrative overview of the engineering principles underlying chemical, optical, electrical, mechanical, and computational science to probe, sense, model, and fabricate the TiME. In individual sections, we first summarize the underlying principles, capabilities, and scope of emerging technologies, the breakthrough discoveries enabled by each technology and recent, promising innovations. We provide perspectives on the potential of these advances in answering critical questions about the TiME and its role in various disease and developmental processes. Finally, we present an integrative view that appreciates the major scientific and educational aspects in the study of the TiME.
Collapse
Affiliation(s)
- Rishyashring R. Iyer
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Catherine C. Applegate
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Opeyemi H. Arogundade
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Sushant Bangru
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Ian C. Berg
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Bashar Emon
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Marilyn Porras-Gomez
- Department of Materials Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Pei-Hsuan Hsieh
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yoon Jeong
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yongdeok Kim
- Department of Materials Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hailey J. Knox
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Amir Ostadi Moghaddam
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Carlos A. Renteria
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Craig Richard
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Ashlie Santaliz-Casiano
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Sourya Sengupta
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jason Wang
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Samantha G. Zambuto
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Maria A. Zeballos
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Marcia Pool
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Rohit Bhargava
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Chemical and Biochemical Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- NIH/NIBIB P41 Center for Label-free Imaging and Multiscale Biophotonics (CLIMB), University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - H. Rex Gaskins
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Biomedical and Translational Sciences, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| |
Collapse
|
50
|
Lembo V, Bottegoni G. Systematic Investigation of Dual-Target-Directed Ligands. J Med Chem 2024; 67:10374-10385. [PMID: 38843874 PMCID: PMC11215722 DOI: 10.1021/acs.jmedchem.4c00838] [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: 04/08/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
Multitarget-directed ligands (MTDLs) are compounds rationally designed to affect multiple targets, aiming for a better therapeutic profile. For over 20 years, MTDLs have garnered increasing attention, the idea being that their full potential would have been achieved, thanks to unprecedented target combinations and advanced medicinal chemistry strategies. This study presents a literature mining effort resulting in a data set of dual-target-directed ligands (DTDLs), the fundamental example of MTDLs. We used this data set to evaluate the rationale behind target selection and the chemical novelty of DTDLs targeting specific protein combinations. Our analysis focused on DTDL targets in terms of biological function, disease association, structure, and chemogenomic traits. We also compared DTDLs with single-target compounds. We found that well-known target pathology associations often guide DTDL design, leveraging existing chemical scaffolds and binding pocket similarities. These findings highlight the current state of the field and suggest substantial untapped potential for rational polypharmacology.
Collapse
Affiliation(s)
- Vittorio Lembo
- Department
of Biomolecular Sciences, Università
degli Studi di Urbino Carlo Bo, Piazza Rinascimento 6, 61029 Urbino, Italy
- Computational
and Chemical Biology, Istituto Italiano
di Tecnologia, Via Morego
30, 16163 Genova, Italy
| | - Giovanni Bottegoni
- Department
of Biomolecular Sciences, Università
degli Studi di Urbino Carlo Bo, Piazza Rinascimento 6, 61029 Urbino, Italy
- Institute
of Clinical Sciences, University of Birmingham, Edgbaston, B15 2TT Birmingham, U.K.
| |
Collapse
|