1
|
Bashore FM, Katis VL, Du Y, Sikdar A, Wang D, Bradshaw WJ, Rygiel KA, Leisner TM, Chalk R, Mishra S, Williams CA, Gileadi O, Brennan PE, Wiley JC, Gockley J, Cary GA, Carter GW, Young JE, Pearce KH, Fu H, the Emory-Sage-SGC TREAT-AD Center, Axtman AD. Characterization of covalent inhibitors that disrupt the interaction between the tandem SH2 domains of SYK and FCER1G phospho-ITAM. PLoS One 2024; 19:e0293548. [PMID: 38359047 PMCID: PMC10868801 DOI: 10.1371/journal.pone.0293548] [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: 08/02/2023] [Accepted: 10/15/2023] [Indexed: 02/17/2024] Open
Abstract
RNA sequencing and genetic data support spleen tyrosine kinase (SYK) and high affinity immunoglobulin epsilon receptor subunit gamma (FCER1G) as putative targets to be modulated for Alzheimer's disease (AD) therapy. FCER1G is a component of Fc receptor complexes that contain an immunoreceptor tyrosine-based activation motif (ITAM). SYK interacts with the Fc receptor by binding to doubly phosphorylated ITAM (p-ITAM) via its two tandem SH2 domains (SYK-tSH2). Interaction of the FCER1G p-ITAM with SYK-tSH2 enables SYK activation via phosphorylation. Since SYK activation is reported to exacerbate AD pathology, we hypothesized that disruption of this interaction would be beneficial for AD patients. Herein, we developed biochemical and biophysical assays to enable the discovery of small molecules that perturb the interaction between the FCER1G p-ITAM and SYK-tSH2. We identified two distinct chemotypes using a high-throughput screen (HTS) and orthogonally assessed their binding. Both chemotypes covalently modify SYK-tSH2 and inhibit its interaction with FCER1G p-ITAM, however, these compounds lack selectivity and this limits their utility as chemical tools.
Collapse
Affiliation(s)
- Frances M. Bashore
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Vittorio L. Katis
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, United States of America
- Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - Arunima Sikdar
- Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Dongxue Wang
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, United States of America
- Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - William J. Bradshaw
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Karolina A. Rygiel
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Tina M. Leisner
- Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Rod Chalk
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Swati Mishra
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, United States of America
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - C. Andrew Williams
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, United States of America
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - Opher Gileadi
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Paul E. Brennan
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | | | - Jake Gockley
- Sage Bionetworks, Seattle, WA, United States of America
| | - Gregory A. Cary
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, United States of America
| | - Gregory W. Carter
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, United States of America
| | - Jessica E. Young
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, United States of America
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - Kenneth H. Pearce
- Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, United States of America
- Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, United States of America
| | | | - Alison D. Axtman
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
2
|
Bashore FM, Katis VL, Du Y, Sikdar A, Wang D, Bradshaw WJ, Rygiel KA, Leisner TM, Chalk R, Mishra S, Williams AC, Gileadi O, Brennan PE, Wiley JC, Gockley J, Cary GA, Carter GW, Young JE, Pearce KH, Fu H, Axtman AD. Characterization of covalent inhibitors that disrupt the interaction between the tandem SH2 domains of SYK and FCER1G phospho-ITAM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.28.551026. [PMID: 37547005 PMCID: PMC10402180 DOI: 10.1101/2023.07.28.551026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
RNA sequencing and genetic data support spleen tyrosine kinase (SYK) and high affinity immunoglobulin epsilon receptor subunit gamma (FCER1G) as putative targets to be modulated for Alzheimer's disease (AD) therapy. FCER1G is a component of Fc receptor complexes that contain an immunoreceptor tyrosine-based activation motif (ITAM). SYK interacts with the Fc receptor by binding to doubly phosphorylated ITAM (p-ITAM) via its two tandem SH2 domains (SYK-tSH2). Interaction of the FCER1G p-ITAM with SYK-tSH2 enables SYK activation via phosphorylation. Since SYK activation is reported to exacerbate AD pathology, we hypothesized that disruption of this interaction would be beneficial for AD patients. Herein, we developed biochemical and biophysical assays to enable the discovery of small molecules that perturb the interaction between the FCER1G p-ITAM and SYK-tSH2. We identified two distinct chemotypes using a high-throughput screen (HTS) and orthogonally assessed their binding. Both chemotypes covalently modify SYK-tSH2 and inhibit its interaction with FCER1G p-ITAM.
Collapse
Affiliation(s)
- Frances M Bashore
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, University of North Carolina, Chapel Hill, NC, USA
| | - Vittorio L Katis
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, USA
| | - Arunima Sikdar
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, University of North Carolina, Chapel Hill, NC, USA
| | - Dongxue Wang
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, USA
| | - William J Bradshaw
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | - Karolina A Rygiel
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | - Tina M Leisner
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, University of North Carolina, Chapel Hill, NC, USA
| | - Rod Chalk
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | | | | | - Opher Gileadi
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
- Current address: Structural Genomics Consortium, Department of Medicine, Karolinska Hospital and Karolinska Institute, 171 76 Stockholm, Sweden
| | - Paul E Brennan
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | | | | | | | | | | | - Kenneth H Pearce
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, University of North Carolina, Chapel Hill, NC, USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, USA
| | - Alison D Axtman
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
3
|
Agarwal P, Huckle J, Newman J, Reid DL. Trends in small molecule drug properties: A developability molecule assessment perspective. Drug Discov Today 2022; 27:103366. [PMID: 36122862 DOI: 10.1016/j.drudis.2022.103366] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/10/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022]
Abstract
Developability molecule assessment is a key interfacial capability across the biopharmaceutical industry, screening and staging molecules discovered by medicinal chemists for successful chemistry manufacturing controls (CMC) development and launch. The breadth of responsibility and expertise such teams possess puts them in a unique position to understand the impact of the physicochemical properties of a drug during its initial discovery and subsequent development. However, most of the publications describing trends in physicochemical properties are written from a medicinal chemistry perspective with the aim to identify molecules with better ADMET profiles that are either lead-like or drug-like, failing to describe the impact these properties have on CMC development. To systematically uncover knowledge obtained from recent trends in physicochemical properties and the corresponding impact on CMC development, a comprehensive analysis was conducted on molecules in the drug repurposing hub dataset. The only physicochemical property that seems to have been preserved in FDA-approved oral molecules over the decades (1900-2020) is a constant H-bond donor count, highlighting the importance this property has on cell permeability and lattice energy. Pharmaceutical attrition analysis suggests that partition-distribution coefficient, H-bond acceptors, polar surface area and the fraction of sp3 carbons are properties that are associated with compound attrition. Looking at pharmaceutical attrition asynchronously with the temporal analysis of FDA-approved oral molecules highlights the opposing trends, risks and diminishing effects some of these physiochemical properties (cLogP, cLogD and Fsp3) have on describing compound attrition during the past decade. Trellising the dataset by target class suggests that certain formulation and drug delivery strategies can be anticipated or put into place based on target class of a molecule. For example, molecules binding to nuclear hormone receptors are amenable to lipid-based drug delivery systems with proven commercial success. Although the poor solubility of kinase inhibitors is a combination of hydrophobicity (due to aromaticity) required to bind to its target and high lattice energy (melting point), they are a challenging target class to formulate. The influence of drug targets on physicochemical properties and the temporal nature of these properties is highlighted when comparing molecules in the drug repurposing dataset to those developed at Amgen. An improved understanding of the impact of molecular properties on performance attributes can accelerate decisions and facilitate risk assessments during candidate selection and development.
Collapse
Affiliation(s)
- Prashant Agarwal
- Drug Product Technologies, Process Development, Amgen, One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | - James Huckle
- Drug Product Technologies, Process Development, Amgen, One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Jake Newman
- Drug Product Technologies, Process Development, Amgen, One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Darren L Reid
- Drug Product Technologies, Process Development, Amgen, 360 Binney St, Cambridge, MA 02142, USA.
| |
Collapse
|
4
|
Bhanot A, Sundriyal S. Physicochemical Profiling and Comparison of Research Antiplasmodials and Advanced Stage Antimalarials with Oral Drugs. ACS OMEGA 2021; 6:6424-6437. [PMID: 33718733 PMCID: PMC7948433 DOI: 10.1021/acsomega.1c00104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/18/2021] [Indexed: 06/12/2023]
Abstract
To understand the property space of antimalarials, we collated a large dataset of research antiplasmodial (RAP) molecules with known in vitro potencies and advanced stage antimalarials (ASAMs) with established oral bioavailability. While RAP molecules are "non-druglike", ASAM molecules display properties closer to Lipinski's and Veber's thresholds. Comparison within the different potency groups of RAP molecules indicates that the in vitro potency is positively correlated to the molecular weight, the calculated octanol-water partition coefficient (clog P), aromatic ring counts (#Ar), and hydrogen bond acceptors. Despite both categories being bioavailable, the ASAM molecules are relatively larger and more lipophilic, have a lower polar surface area, and possess a higher count of heteroaromatic rings than oral drugs. Also, antimalarials are found to have a higher proportion of aromatic (#ArN) and basic nitrogen (#BaN) counts, features implicitly used in the design of antimalarial molecules but not well studied hitherto. We also propose using descriptors scaled by the sum of #ArN and #BaN (SBAN) to define an antimalarial property space. Together, these results may have important applications in the identification and optimization of future antimalarials.
Collapse
Affiliation(s)
- Amritansh Bhanot
- Department of Pharmacy, Birla
Institute of Technology and Science Pilani, Pilani Campus,
Vidya Vihar, Pilani, Rajasthan 333 031, India
| | - Sandeep Sundriyal
- Department of Pharmacy, Birla
Institute of Technology and Science Pilani, Pilani Campus,
Vidya Vihar, Pilani, Rajasthan 333 031, India
| |
Collapse
|
5
|
Fu L, Yang ZY, Yang ZJ, Yin MZ, Lu AP, Chen X, Liu S, Hou TJ, Cao DS. QSAR-assisted-MMPA to expand chemical transformation space for lead optimization. Brief Bioinform 2021; 22:6071857. [PMID: 33418563 DOI: 10.1093/bib/bbaa374] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/25/2020] [Accepted: 11/25/2020] [Indexed: 11/13/2022] Open
Abstract
Matched molecular pairs analysis (MMPA) has become a powerful tool for automatically and systematically identifying medicinal chemistry transformations from compound/property datasets. However, accurate determination of matched molecular pair (MMP) transformations largely depend on the size and quality of existing experimental data. Lack of high-quality experimental data heavily hampers the extraction of more effective medicinal chemistry knowledge. Here, we developed a new strategy called quantitative structure-activity relationship (QSAR)-assisted-MMPA to expand the number of chemical transformations and took the logD7.4 property endpoint as an example to demonstrate the reliability of the new method. A reliable logD7.4 consensus prediction model was firstly established, and its applicability domain was strictly assessed. By applying the reliable logD7.4 prediction model to screen two chemical databases, we obtained more high-quality logD7.4 data by defining a strict applicability domain threshold. Then, MMPA was performed on the predicted data and experimental data to derive more chemical rules. To validate the reliability of the chemical rules, we compared the magnitude and directionality of the property changes of the predicted rules with those of the measured rules. Then, we compared the novel chemical rules generated by our proposed approach with the published chemical rules, and found that the magnitude and directionality of the property changes were consistent, indicating that the proposed QSAR-assisted-MMPA approach has the potential to enrich the collection of rule types or even identify completely novel rules. Finally, we found that the number of the MMP rules derived from the experimental data could be amplified by the predicted data, which is helpful for us to analyze the medicinal chemical rules in local chemical environment. In summary, the proposed QSAR-assisted-MMPA approach could be regarded as a very promising strategy to expand the chemical transformation space for lead optimization, especially when no enough experimental data can support MMPA.
Collapse
Affiliation(s)
- Li Fu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China.,Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
| | - Zi-Yi Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
| | - Zhi-Jiang Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
| | - Ming-Zhu Yin
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
| | - Ai-Ping Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, P. R China
| | - Xiang Chen
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China
| | - Shao Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
| | - Ting-Jun Hou
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China
| | - Dong-Sheng Cao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China.,Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China.,Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, P. R China
| |
Collapse
|
6
|
Sivaraman G, Jackson NE, Sanchez-Lengeling B, Vázquez-Mayagoitia Á, Aspuru-Guzik A, Vishwanath V, de Pablo JJ. A machine learning workflow for molecular analysis: application to melting points. MACHINE LEARNING: SCIENCE AND TECHNOLOGY 2020. [DOI: 10.1088/2632-2153/ab8aa3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Abstract
Computational tools encompassing integrated molecular prediction, analysis, and generation are key for molecular design in a variety of critical applications. In this work, we develop a workflow for molecular analysis (MOLAN) that integrates an ensemble of supervised and unsupervised machine learning techniques to analyze molecular data sets. The MOLAN workflow combines molecular featurization, clustering algorithms, uncertainty analysis, low-bias dataset construction, high-performance regression models, graph-based molecular embeddings and attribution, and a semi-supervised variational autoencoder based on the novel SELFIES representation to enable molecular design. We demonstrate the utility of the MOLAN workflow in the context of a challenging multi-molecule property prediction problem: the determination of melting points solely from single molecule structure. This application serves as a case study for how to employ the MOLAN workflow in the context of molecular property prediction.
Collapse
|
7
|
Tinworth CP, Young RJ. Facts, Patterns, and Principles in Drug Discovery: Appraising the Rule of 5 with Measured Physicochemical Data. J Med Chem 2020; 63:10091-10108. [PMID: 32324397 DOI: 10.1021/acs.jmedchem.9b01596] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The rule of 5 was designed to estimate the likelihood of poor absorption or permeation, noting the impact of poor solubility. This Perspective explores the impact of various physicochemical descriptors and contemporary lipophilicity measurements on permeability and solubility, showing that the distribution coefficient log D7.4 (rather than log P) is the most impactful parameter. Molecular weight, almost invariably the defining characteristic of "beyond the rule of 5" compounds, has little impact on solubility when log D7.4 measurements and aromaticity are considered. Predicting permeation is more complex, given passive and carrier transport mechanisms; however, notable patterns of behavior are apparent, giving insight even "beyond the rule of 5". Recommended best practices should involve using the facts (measurements) and the patterns they reveal to establish informative principles rather than fastidious rules.
Collapse
Affiliation(s)
- Christopher P Tinworth
- Medicinal Sciences and Technology, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Robert J Young
- Medicinal Sciences and Technology, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K.,Blue Burgundy Ltd., Bedford, Bedfordshire MK45 2AD, U.K
| |
Collapse
|
8
|
Fine JA, Rajasekar AA, Jethava KP, Chopra G. Spectral deep learning for prediction and prospective validation of functional groups. Chem Sci 2020; 11:4618-4630. [PMID: 34122917 PMCID: PMC8152587 DOI: 10.1039/c9sc06240h] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/13/2020] [Indexed: 01/06/2023] Open
Abstract
State-of-the-art identification of the functional groups present in an unknown chemical entity requires the expertise of a skilled spectroscopist to analyse and interpret Fourier transform infra-red (FTIR), mass spectroscopy (MS) and/or nuclear magnetic resonance (NMR) data. This process can be time-consuming and error-prone, especially for complex chemical entities that are poorly characterised in the literature, or inefficient to use with synthetic robots producing molecules at an accelerated rate. Herein, we introduce a fast, multi-label deep neural network for accurately identifying all the functional groups of unknown compounds using a combination of FTIR and MS spectra. We do not use any database, pre-established rules, procedures, or peak-matching methods. Our trained neural network reveals patterns typically used by human chemists to identify standard groups. Finally, we experimentally validated our neural network, trained on single compounds, to predict functional groups in compound mixtures. Our methodology showcases practical utility for future use in autonomous analytical detection.
Collapse
Affiliation(s)
- Jonathan A Fine
- Department of Chemistry, Purdue University 560 Oval Drive West Lafayette IN 47907 USA
| | - Anand A Rajasekar
- Department of Biological Engineering, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras Chennai 600036 India
| | - Krupal P Jethava
- Department of Chemistry, Purdue University 560 Oval Drive West Lafayette IN 47907 USA
| | - Gaurav Chopra
- Department of Chemistry, Purdue University 560 Oval Drive West Lafayette IN 47907 USA
| |
Collapse
|
9
|
Fu L, Liu L, Yang ZJ, Li P, Ding JJ, Yun YH, Lu AP, Hou TJ, Cao DS. Systematic Modeling of log D7.4 Based on Ensemble Machine Learning, Group Contribution, and Matched Molecular Pair Analysis. J Chem Inf Model 2019; 60:63-76. [DOI: 10.1021/acs.jcim.9b00718] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Li Fu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
| | - Lu Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
| | - Zhi-Jiang Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
| | - Pan Li
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, P. R. China
| | - Jun-Jie Ding
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, P. R. China
| | - Yong-Huan Yun
- College of Food Science and Engineering, Hainan University, Haikou 570228, P. R. China
| | - Ai-Ping Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, P. R. China
| | - Ting-Jun Hou
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China
| | - Dong-Sheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, P. R. China
| |
Collapse
|
10
|
Karlov DS, Sosnin S, Tetko IV, Fedorov MV. Chemical space exploration guided by deep neural networks. RSC Adv 2019; 9:5151-5157. [PMID: 35514634 PMCID: PMC9060647 DOI: 10.1039/c8ra10182e] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/29/2019] [Indexed: 11/21/2022] Open
Abstract
A parametric t-SNE approach based on deep feed-forward neural networks was applied to the chemical space visualization problem. It is able to retain more information than certain dimensionality reduction techniques used for this purpose (principal component analysis (PCA), multidimensional scaling (MDS)). The applicability of this method to some chemical space navigation tasks (activity cliffs and activity landscapes identification) is discussed. We created a simple web tool to illustrate our work (http://space.syntelly.com). A parametric t-SNE approach based on deep feed-forward neural networks was applied to the chemical space visualization problem.![]()
Collapse
Affiliation(s)
- Dmitry S. Karlov
- Skolkovo Institute of Science and Technology
- Skolkovo Innovation Center
- Moscow 143026
- Russia
| | - Sergey Sosnin
- Skolkovo Institute of Science and Technology
- Skolkovo Innovation Center
- Moscow 143026
- Russia
- Syntelly LLC
| | - Igor V. Tetko
- Helmholtz Zentrum München – Research Center for Environmental Health (GmbH)
- Institute of Structural Biology
- Germany
- BIGCHEM GmbH
- Germany
| | - Maxim V. Fedorov
- Skolkovo Institute of Science and Technology
- Skolkovo Innovation Center
- Moscow 143026
- Russia
- Syntelly LLC
| |
Collapse
|
11
|
Fournier JF, Clary L, Chambon S, Dumais L, Harris CS, Millois C, Pierre R, Talano S, Thoreau É, Aubert J, Aurelly M, Bouix-Peter C, Brethon A, Chantalat L, Christin O, Comino C, El-Bazbouz G, Ghilini AL, Isabet T, Lardy C, Luzy AP, Mathieu C, Mebrouk K, Orfila D, Pascau J, Reverse K, Roche D, Rodeschini V, Hennequin LF. Rational Drug Design of Topically Administered Caspase 1 Inhibitors for the Treatment of Inflammatory Acne. J Med Chem 2018; 61:4030-4051. [DOI: 10.1021/acs.jmedchem.8b00067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jean-François Fournier
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Laurence Clary
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Sandrine Chambon
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Laurence Dumais
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Craig Steven Harris
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Corinne Millois
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Romain Pierre
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Sandrine Talano
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Étienne Thoreau
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Jérome Aubert
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Michèle Aurelly
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Claire Bouix-Peter
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Anne Brethon
- Edelris, 115 Avenue Lacassagne, 69003 Lyon, France
| | - Laurent Chantalat
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Olivier Christin
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Catherine Comino
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Ghizlane El-Bazbouz
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Anne-Laurence Ghilini
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Tatiana Isabet
- Synchrotron Soleil, L’Orme des Merisiers, Saint-Aubin, BP 48, 91192 Gif-sur-Yvette Cedex, France
| | - Claude Lardy
- Edelris, 115 Avenue Lacassagne, 69003 Lyon, France
| | - Anne-Pascale Luzy
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Céline Mathieu
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Kenny Mebrouk
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Danielle Orfila
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Jonathan Pascau
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Kevin Reverse
- Nestlé Skin Health R&D, 2400 Route des Colles, BP 87, 06902 Sophia-Antipolis Cedex, France
| | - Didier Roche
- Edelris, 115 Avenue Lacassagne, 69003 Lyon, France
| | | | | |
Collapse
|
12
|
Withnall M, Chen H, Tetko IV. Matched Molecular Pair Analysis on Large Melting Point Datasets: A Big Data Perspective. ChemMedChem 2018; 13:599-606. [PMID: 28650584 PMCID: PMC5900986 DOI: 10.1002/cmdc.201700303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 06/26/2017] [Indexed: 11/11/2022]
Abstract
A matched molecular pair (MMP) analysis was used to examine the change in melting point (MP) between pairs of similar molecules in a set of ∼275k compounds. We found many cases in which the change in MP (ΔMP) of compounds correlates with changes in functional groups. In line with the results of a previous study, correlations between ΔMP and simple molecular descriptors, such as the number of hydrogen bond donors, were identified. In using a larger dataset, covering a wider chemical space and range of melting points, we observed that this method remains stable and scales well with larger datasets. This MMP-based method could find use as a simple privacy-preserving technique to analyze large proprietary databases and share findings between participating research groups.
Collapse
Affiliation(s)
- Michael Withnall
- Helmholtz Zentrum München—German Research Center for Environmental Health, GmbHInstitute of Structural BiologyNeuherbergGermany
| | - Hongming Chen
- External Sciences, Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D GothenburgMölndal43183Sweden
| | - Igor V. Tetko
- Helmholtz Zentrum München—German Research Center for Environmental Health, GmbHInstitute of Structural BiologyNeuherbergGermany
- BIGCHEM GmbHIngolstädter Landstraße 1, b. 60w85764NeuherbergGermany
- Institute of Structural Biology, Helmholtz Zentrum München—German Research Center for Environmental Health, GmbHIngolstädter Landstraße 185764NeuherbergGermany
| |
Collapse
|