1
|
Cozac R, Hasic H, Choong JJ, Richard V, Beheshti L, Froehlich C, Koyama T, Matsumoto S, Kojima R, Iwata H, Hasegawa A, Otsuka T, Okuno Y. kMoL: an open-source machine and federated learning library for drug discovery. J Cheminform 2025; 17:22. [PMID: 40001146 PMCID: PMC11854109 DOI: 10.1186/s13321-025-00967-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Machine learning is quickly becoming integral to drug discovery pipelines, particularly quantitative structure-activity relationship (QSAR) and absorption, distribution, metabolism, and excretion (ADME) tasks. Graph Convolutional Network (GCN) models have proven especially promising due to their inherent ability to model molecular structures using graph-based representations. However, maximizing the potential of such models in practice is challenging, as companies prioritize data privacy and security over collaboration initiatives to improve model performance and robustness. kMoL is an open-source machine learning library with integrated federated learning capabilities developed to address such challenges. Its key features include state-of-the-art model architectures, Bayesian optimization, explainability, and federated learning mechanisms. It demonstrates extensive customization possibilities, advanced security features, straightforward implementation of user-specific models, and high adaptability to custom datasets without additional programming requirements. kMoL is evaluated through locally trained benchmark settings and distributed federated learning experiments using various datasets to assess the features and flexibility of the library, as well as the ability to facilitate fast and practical experimentation. Additionally, results of these experiments provide further insights into the performance trade-offs associated with federated learning strategies, presenting valuable guidance for deploying machine learning models in a privacy-preserving manner within drug discovery pipelines. kMoL is available on GitHub at https://github.com/elix-tech/kmol .Scientific contribution The primary scientific contribution of this research project is the introduction and evaluation of kMoL, an open-source machine learning library with integrated federated learning capabilities. By demonstrating advanced customization and security capabilities without additional programming requirements, kMoL represents an accessible yet secure open-source platform for collaborative drug discovery projects. Additionally, the experiment results provide further insights into the performance trade-offs associated with federated learning strategies, presenting valuable guidance for deploying machine learning models in a privacy-preserving manner within drug discovery pipelines.
Collapse
Affiliation(s)
- Romeo Cozac
- Elix, Inc., 8-34 Yonbancho, Chiyoda-ku, Tokyo, 102-0081, Japan.
| | - Haris Hasic
- Elix, Inc., 8-34 Yonbancho, Chiyoda-ku, Tokyo, 102-0081, Japan
| | - Jun Jin Choong
- Elix, Inc., 8-34 Yonbancho, Chiyoda-ku, Tokyo, 102-0081, Japan
| | - Vincent Richard
- Elix, Inc., 8-34 Yonbancho, Chiyoda-ku, Tokyo, 102-0081, Japan
| | - Loic Beheshti
- Elix, Inc., 8-34 Yonbancho, Chiyoda-ku, Tokyo, 102-0081, Japan
| | | | - Takuto Koyama
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shigeyuki Matsumoto
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ryosuke Kojima
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroaki Iwata
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Aki Hasegawa
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takao Otsuka
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yasushi Okuno
- Graduate School of Medicine, Kyoto University, Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
2
|
Han Z, Xia Z, Xia J, Tetko IV, Wu S. The state-of-the-art machine learning model for plasma protein binding prediction: Computational modeling with OCHEM and experimental validation. Eur J Pharm Sci 2025; 204:106946. [PMID: 39490636 DOI: 10.1016/j.ejps.2024.106946] [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: 07/30/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Plasma protein binding (PPB) is closely related to pharmacokinetics, pharmacodynamics and drug toxicity. Existing models for predicting PPB often suffer from low prediction accuracy and poor interpretability, especially for high PPB compounds, and are most often not experimentally validated. Here, we carried out a strict data curation protocol, and applied consensus modeling to obtain a model with a coefficient of determination of 0.90 and 0.91 on the training set and the test set, respectively. This model (available on the OCHEM platform https://ochem.eu/article/29) was further retrospectively validated for a set of 63 poly-fluorinated molecules and prospectively validated for a set of 25 highly diverse compounds, and its performance for both these sets was superior to that of the other previously reported models. Furthermore, we identified the physicochemical and structural characteristics of high and low PPB molecules for further structural optimization. Finally, we provide practical and detailed recommendations for structural optimization to decrease PPB binding of lead compounds.
Collapse
Affiliation(s)
- Zunsheng Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhonghua Xia
- Institute of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Jie Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Igor V Tetko
- Institute of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany; BIGCHEM GmbH, Valerystr. 49, 85716 Unterschleißheim, Germany.
| | - Song Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
3
|
Tanaka T, Katayama T, Imai T. Predicting the effects of drugs and unveiling their mechanisms of action using an interpretable pharmacodynamic mechanism knowledge graph (IPM-KG). Comput Biol Med 2025; 184:109419. [PMID: 39556916 DOI: 10.1016/j.compbiomed.2024.109419] [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: 03/11/2024] [Revised: 10/18/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Multiple studies have aimed to consolidate drug-related data and predict drug effects. However, most of these studies have focused on integrating diverse data through correlation rather than representing them based on the pharmacodynamic mechanism of action (MOA). It is thus crucial to obtain interpretability to validate prediction results. In this study, we propose a novel framework to construct knowledge graphs that represent pharmacodynamic MOA, predict drug effects, and derive conceivable mechanistic pathways. METHODS AND RESULTS We constructed an interpretable pharmacodynamic mechanism knowledge graph (IPM-KG) by integrating various existing databases and combining them with the approach of this study to automatically fill in the missing data. This yielded a knowledge graph comprising 1455 drugs and 2547 diseases. Additionally, a graph neural network (GNN)-based approach was used to predict therapeutic medication and indication, which outperformed previous approaches that relied on correlation-based knowledge graphs lacking pharmacodynamic MOA representations. Furthermore, we proposed and assessed a method to interpret pharmacodynamic MOA using gene perturbation data. This feasibility study demonstrated the successful derivation of an accurate mechanism in approximately 50 % of cases. Additionally, it facilitated the identification of candidate drugs, which are currently unapproved but exhibit potential for drug repositioning, and their mechanisms of action. CONCLUSIONS This framework not only enables the derivation of highly accurate "drug-indication" predictions but also offers a basic mechanistic understanding, thereby facilitating future drug repositioning efforts.
Collapse
Affiliation(s)
- Tatsuya Tanaka
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiaki Katayama
- Bio Data Science Initiative and Database Center for Life Science, Joint Support-Center for Data Science Research, Research Organization of Information and Systems, Chiba, Japan
| | - Takeshi Imai
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
4
|
Kwon JH, Han JY, Kim M, Kim SK, Lee DK, Kim MG. Prediction of human pharmacokinetic parameters incorporating SMILES information. Arch Pharm Res 2024; 47:914-923. [PMID: 39589671 DOI: 10.1007/s12272-024-01520-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 11/16/2024] [Indexed: 11/27/2024]
Abstract
This study aimed to develop a model incorporating natural language processing analysis for the simplified molecular-input line-entry system (SMILES) to predict clearance (CL) and volume of distribution at steady state (Vd,ss) in humans. The construction of CL and Vd,ss prediction models involved data from 435 to 439 compounds, respectively. In machine learning, features such as animal pharmacokinetic data, in vitro experimental data, molecular descriptors, and SMILES were utilized, with XGBoost employed as the algorithm. The ChemBERTa model was used to analyze substance SMILES, and the last hidden layer embedding of ChemBERTa was examined as a feature. The model was evaluated using geometric mean fold error (GMFE), r2, root mean squared error (RMSE), and accuracy within 2- and 3-fold error. The model demonstrated optimal performance for CL prediction when incorporating animal pharmacokinetic data, in vitro experimental data, and SMILES as features, yielding a GMFE of 1.768, an r2 of 0.528, an RMSE of 0.788, with accuracies within 2-fold and 3-fold error reaching 75.8% and 81.8%, respectively. The model's performance in Vd,ss prediction was optimized by leveraging animal pharmacokinetic data and in vitro experimental data as features, yielding a GMFE of 1.401, an r2 of 0.902, an RMSE of 0.413, with accuracies within 2-fold and 3-fold error reaching 93.8% and 100%, respectively. This study has developed a highly predictive model for CL and Vd,ss. Specifically, incorporating SMILES information into the model has predictive power for CL.
Collapse
Affiliation(s)
- Jae-Hee Kwon
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Ja-Young Han
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Minjung Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seong Kyung Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Dong-Kyu Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Myeong Gyu Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
- College of Pharmacy, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
5
|
Baidya ATK, Goswami AK, Das B, Darreh-Shori T, Kumar R. AI-Enabled Ultra-large Virtual Screening Identifies Potential Inhibitors of Choline Acetyltransferase for Theranostic Purposes. ACS Chem Neurosci 2024; 15:4156-4170. [PMID: 39481020 DOI: 10.1021/acschemneuro.4c00361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Abstract
Alzheimer's disease (AD) and related dementias are among the primary neurological disorders and call for the urgent need for early-stage diagnosis to gain an upper edge in therapeutic intervention and increase the overall success rate. Choline acetyltransferase (ChAT) is the key acetylcholine (ACh) biosynthesizing enzyme and a legitimate target for the development of biomarkers for early-stage diagnosis and monitoring of therapeutic responses. It is also a theranostic target for tackling colon and lung cancers, where overexpression of non-neuronal ChAT leads to the production of acetylcholine, which acts as an autocrine growth factor for cancer cells. Theranostics is a hybrid of diagnostics and therapeutics that can be used to locate cancer cells using radiotracers and kill them without affecting other healthy tissues. Traditional virtual screening protocols have a lot of limitations; given the current rate of chemical database expansion exceeding billions, much faster screening protocols are required. Deep docking (DD) is one such platform that leverages the power of deep neural network (DNN)-based virtual screening, empowering researchers to dock billions of molecules in a speedy, yet explicit manner. Here, we have screened 1.3 billion compounds library from the ZINC20 database, identifying the best-performing hits. With each iteration run where the first iteration gave ∼116 million hits, the second iteration gave ∼3.7 million hits, and the final third iteration gave 168,447 hits from which further refinement gave us the top 5 compounds as potential ChAT inhibitors. The discovery of novel ChAT inhibitors will enable researchers to develop new probes that can be used as novel theranostic agents against cancer and as early-stage diagnostics for the onset of AD, for timely therapeutic intervention to halt the further progression of AD.
Collapse
Affiliation(s)
- Anurag T K Baidya
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India
| | - Abhinav Kumar Goswami
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India
| | - Bhanuranjan Das
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India
| | - Taher Darreh-Shori
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Rajnish Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India
| |
Collapse
|
6
|
Rich AS, Chan YH, Birnbaum B, Haider K, Haimson J, Hale M, Han Y, Hickman W, Hoeflich KP, Ortwine D, Özen A, Belanger DB. Machine Learning ADME Models in Practice: Four Guidelines from a Successful Lead Optimization Case Study. ACS Med Chem Lett 2024; 15:1169-1173. [PMID: 39140048 PMCID: PMC11318014 DOI: 10.1021/acsmedchemlett.4c00290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 06/26/2024] [Indexed: 08/15/2024] Open
Abstract
Optimization of the ADME properties and pharmacokinetic (PK) profile of compounds is one of the critical activities in any medicinal chemistry campaign to discover a future clinical candidate. Finding ways to expedite the process to address ADME/PK shortcomings and reduce the number of compounds to synthesize is highly valuable. This article provides practical guidelines and a case study on the use of ML ADME models to guide compound design in small molecule lead optimization. These guidelines highlight that ML models cannot have an impact in a vacuum: they help advance a program when they have the trust of users, are tuned to the needs of the program, and are integrated into decision-making processes in a way that complements and augments the expertise of chemists.
Collapse
Affiliation(s)
- Alexander S. Rich
- Inductive
Bio, Inc., 550 Vanderbilt
Ave, #730, Brooklyn, New
York 11238, United
States
| | - Yvonne H. Chan
- Nested
Therapeutics, 1030 Mass
Ave, Suite 410, Cambridge, Massachusetts 02138, United States
| | - Benjamin Birnbaum
- Inductive
Bio, Inc., 550 Vanderbilt
Ave, #730, Brooklyn, New
York 11238, United
States
| | - Kamran Haider
- Nested
Therapeutics, 1030 Mass
Ave, Suite 410, Cambridge, Massachusetts 02138, United States
| | - Joshua Haimson
- Inductive
Bio, Inc., 550 Vanderbilt
Ave, #730, Brooklyn, New
York 11238, United
States
| | - Michael Hale
- Nested
Therapeutics, 1030 Mass
Ave, Suite 410, Cambridge, Massachusetts 02138, United States
| | - Yongxin Han
- Nested
Therapeutics, 1030 Mass
Ave, Suite 410, Cambridge, Massachusetts 02138, United States
| | - William Hickman
- Inductive
Bio, Inc., 550 Vanderbilt
Ave, #730, Brooklyn, New
York 11238, United
States
| | - Klaus P. Hoeflich
- Nested
Therapeutics, 1030 Mass
Ave, Suite 410, Cambridge, Massachusetts 02138, United States
| | - Daniel Ortwine
- Nested
Therapeutics, 1030 Mass
Ave, Suite 410, Cambridge, Massachusetts 02138, United States
| | - Ayşegül Özen
- Nested
Therapeutics, 1030 Mass
Ave, Suite 410, Cambridge, Massachusetts 02138, United States
| | - David B. Belanger
- Nested
Therapeutics, 1030 Mass
Ave, Suite 410, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
7
|
Okada M, Tran TTT. Effect of chronic administration of ostruthin on depression-like behavior in chronically stressed mice. IBRO Neurosci Rep 2024; 16:622-628. [PMID: 38832088 PMCID: PMC11144753 DOI: 10.1016/j.ibneur.2024.05.009] [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: 02/29/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
We have previously shown that a single dose of a TREK-1 channel activator, ostruthin, exhibited antidepressant and anxiolytic effects in acute behavioral test models in mice. To assess the potential clinical application, it is essential to evaluate the effects of long-term administration of ostruthin in a chronically stressed mouse model, which is considered to be similar to the clinical condition of major depression in humans. Here, we tested the effects of a single and a 7-day administration of ostruthin on mice that were subjected to chronic unpredictable mild stress (CUMS). A single administration of ostruthin showed antidepressive effects in the tail suspension and forced swim tests of CUMS-treated mice. Unexpectedly, the 7-day administration exhibited only insignificant antidepressive and anxiolytic effects. The 7-day regimen did not affect food intake or body-weight gain, suggesting the absence of apparent cytotoxicity. The mice receiving the 7-day administration had significantly lower blood concentrations of ostruthin compared to those receiving a single dose, suggesting an upregulation of drug-metabolizing activities. These findings suggest that there is a need for stable TREK-1 channel activators that are not affected by drug metabolism.
Collapse
Affiliation(s)
- Masayoshi Okada
- Department of Medical LifeScience, College of Life Science, Kurashiki University of Science and the Arts, Kurashiki, Okayama 712-8505, Japan
| | - Thi Thu Thuy Tran
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
| |
Collapse
|
8
|
Baillache DJ, Valero T, Lorente-Macías Á, Bennett DJ, Elliott RJR, Carragher NO, Unciti-Broceta A. Discovery of pyrazolopyrimidines that selectively inhibit CSF-1R kinase by iterative design, synthesis and screening against glioblastoma cells. RSC Med Chem 2023; 14:2611-2624. [PMID: 38099057 PMCID: PMC10718585 DOI: 10.1039/d3md00454f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/09/2023] [Indexed: 12/17/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of brain cancer in adults, with an average life expectancy under treatment of approx. 15 months. GBM is characterised by a complex set of genetic alterations that results in significant disruption of receptor tyrosine kinase (RTK) signaling. We report here an exploration of the pyrazolo[3,4-d]pyrimidine scaffold in search for antiproliferative compounds directed to GBM treatment. Small compound libraries were synthesised and screened against GBM cells to build up structure-antiproliferative activity-relationships (SAARs) and inform further rounds of design, synthesis and screening. 76 novel compounds were generated through this iterative process that found low micromolar potencies against selected GBM lines, including patient-derived stem cells. Phenomics analysis demonstrated preferential activity against glioma cells of the mesenchymal subtype, whereas kinome screening identified colony stimulating factor-1 receptor (CSF-1R) as the lead's target, a RTK implicated in the tumourigenesis and progression of different cancers and the immunoregulation of the GBM microenvironment.
Collapse
Affiliation(s)
- Daniel J Baillache
- Edinburgh Cancer Research, Institute of Genetics & Cancer, University of Edinburgh Crewe Road South Edinburgh EH4 2XR UK
- Cancer Research UK Scotland Centre UK
| | - Teresa Valero
- Edinburgh Cancer Research, Institute of Genetics & Cancer, University of Edinburgh Crewe Road South Edinburgh EH4 2XR UK
- Cancer Research UK Scotland Centre UK
| | - Álvaro Lorente-Macías
- Edinburgh Cancer Research, Institute of Genetics & Cancer, University of Edinburgh Crewe Road South Edinburgh EH4 2XR UK
- Cancer Research UK Scotland Centre UK
| | | | - Richard J R Elliott
- Edinburgh Cancer Research, Institute of Genetics & Cancer, University of Edinburgh Crewe Road South Edinburgh EH4 2XR UK
- Cancer Research UK Scotland Centre UK
| | - Neil O Carragher
- Edinburgh Cancer Research, Institute of Genetics & Cancer, University of Edinburgh Crewe Road South Edinburgh EH4 2XR UK
- Cancer Research UK Scotland Centre UK
| | - Asier Unciti-Broceta
- Edinburgh Cancer Research, Institute of Genetics & Cancer, University of Edinburgh Crewe Road South Edinburgh EH4 2XR UK
- Cancer Research UK Scotland Centre UK
| |
Collapse
|
9
|
Komura H, Watanabe R, Mizuguchi K. The Trends and Future Prospective of In Silico Models from the Viewpoint of ADME Evaluation in Drug Discovery. Pharmaceutics 2023; 15:2619. [PMID: 38004597 PMCID: PMC10675155 DOI: 10.3390/pharmaceutics15112619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Drug discovery and development are aimed at identifying new chemical molecular entities (NCEs) with desirable pharmacokinetic profiles for high therapeutic efficacy. The plasma concentrations of NCEs are a biomarker of their efficacy and are governed by pharmacokinetic processes such as absorption, distribution, metabolism, and excretion (ADME). Poor ADME properties of NCEs are a major cause of attrition in drug development. ADME screening is used to identify and optimize lead compounds in the drug discovery process. Computational models predicting ADME properties have been developed with evolving model-building technologies from a simplified relationship between ADME endpoints and physicochemical properties to machine learning, including support vector machines, random forests, and convolution neural networks. Recently, in the field of in silico ADME research, there has been a shift toward evaluating the in vivo parameters or plasma concentrations of NCEs instead of using predictive results to guide chemical structure design. Another research hotspot is the establishment of a computational prediction platform to strengthen academic drug discovery. Bioinformatics projects have produced a series of in silico ADME models using free software and open-access databases. In this review, we introduce prediction models for various ADME parameters and discuss the currently available academic drug discovery platforms.
Collapse
Affiliation(s)
- Hiroshi Komura
- University Research Administration Center, Osaka Metropolitan University, 1-2-7 Asahimachi, Abeno-ku, Osaka 545-0051, Osaka, Japan
| | - Reiko Watanabe
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita 565-0871, Osaka, Japan; (R.W.); (K.M.)
- Artificial Intelligence Centre for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN), 3-17 Senrioka-shinmachi, Settu 566-0002, Osaka, Japan
| | - Kenji Mizuguchi
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita 565-0871, Osaka, Japan; (R.W.); (K.M.)
- Artificial Intelligence Centre for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN), 3-17 Senrioka-shinmachi, Settu 566-0002, Osaka, Japan
| |
Collapse
|