1
|
An JP, Wang Y, Munger SD, Tang X. A review on natural sweeteners, sweet taste modulators and bitter masking compounds: structure-activity strategies for the discovery of novel taste molecules. Crit Rev Food Sci Nutr 2024:1-24. [PMID: 38494695 DOI: 10.1080/10408398.2024.2326012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Growing demand for the tasty and healthy food has driven the development of low-calorie sweeteners, sweet taste modulators, and bitter masking compounds originated from natural sources. With the discovery of human taste receptors, increasing numbers of sweet taste modulators have been identified through human taste response and molecular docking techniques. However, the discovery of novel taste-active molecules in nature can be accelerated by using advanced spectrometry technologies based on structure-activity relationships (SARs). SARs explain why structurally similar compounds can elicit similar taste qualities. Given the characterization of structural information from reported data, strategies employing SAR techniques to find structurally similar compounds become an innovative approach to expand knowledge of sweeteners. This review aims to summarize the structural patterns of known natural non-nutritive sweeteners, sweet taste enhancers, and bitter masking compounds. Innovative SAR-based approaches to explore sweetener derivatives are also discussed. Most sweet-tasting flavonoids belong to either the flavanonols or the dihydrochalcones and known bitter masking molecules are flavanones. Based on SAR findings that structural similarities are related to the sensory properties, innovative methodologies described in this paper can be applied to screen and discover the derivatives of taste-active compounds or potential taste modulators.
Collapse
Affiliation(s)
- Jin-Pyo An
- Food Science and Human Nutrition, Citrus Research and Education Center, University of Florida, Lake Alfred, FL, USA
| | - Yu Wang
- Food Science and Human Nutrition, Citrus Research and Education Center, University of Florida, Lake Alfred, FL, USA
| | - Steven D Munger
- Center for Smell and Taste, Department of Pharmacology and Therapeutics, Department of Otolaryngology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Xixuan Tang
- Food Science and Human Nutrition, Citrus Research and Education Center, University of Florida, Lake Alfred, FL, USA
| |
Collapse
|
2
|
Gopal D, Muthuraj R, Balaya RDA, Kanekar S, Ahmed I, Chandrasekaran J. Computational discovery of novel FYN kinase inhibitors: a cheminformatics and machine learning-driven approach to targeted cancer and neurodegenerative therapy. Mol Divers 2024:10.1007/s11030-024-10819-7. [PMID: 38418686 DOI: 10.1007/s11030-024-10819-7] [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: 11/20/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
In this study, we explored the potential of novel inhibitors for FYN kinase, a critical target in cancer and neurodegenerative disorders, by integrating advanced cheminformatics, machine learning, and molecular simulation techniques. Our approach involved analyzing key interactions for FYN inhibition using established multi-kinase inhibitors such as Staurosporine, Dasatinib, and Saracatinib. We utilized ECFP4 circular fingerprints and the t-SNE machine learning algorithm to compare molecular similarities between FDA-approved drugs and known clinical trial inhibitors. This led to the identification of potential inhibitors, including Afatinib, Copanlisib, and Vandetanib. Using the DrugSpaceX platform, we generated a vast library of 72,196 analogues from these leads, which after careful refinement, resulted in 6008 promising candidates. Subsequent clustering identified 48 analogues with significant similarity to known inhibitors. Notably, two candidates derived from Vandetanib, DE27123047 and DE27123035, exhibited strong docking affinities and stable binding in molecular dynamics simulations. These candidates showed high potential as effective FYN kinase inhibitors, as evidenced by MMGBSA calculations and MCE-18 scores exceeding 50. Additionally, our exploration into their molecular architecture revealed potential modification sites on the quinazolin-4-amine scaffold, suggesting opportunities for strategic alterations to enhance activity and optimize ADME properties. Our research is a pioneering effort in drug discovery, unveiling novel candidates for FYN inhibition and demonstrating the efficacy of a multi-layered computational strategy. The molecular insights gained provide a pathway for strategic refinements and future experimental validations, setting a new direction in targeted drug development against diseases involving FYN kinase.
Collapse
Affiliation(s)
- Dhanushya Gopal
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, 600116, India
| | - Rajesh Muthuraj
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, 600116, India
| | | | - Saptami Kanekar
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Iqrar Ahmed
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Dhule, India
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Jaikanth Chandrasekaran
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, 600116, India.
| |
Collapse
|
3
|
Niazi SK, Mariam Z. Recent Advances in Machine-Learning-Based Chemoinformatics: A Comprehensive Review. Int J Mol Sci 2023; 24:11488. [PMID: 37511247 PMCID: PMC10380192 DOI: 10.3390/ijms241411488] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/30/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
In modern drug discovery, the combination of chemoinformatics and quantitative structure-activity relationship (QSAR) modeling has emerged as a formidable alliance, enabling researchers to harness the vast potential of machine learning (ML) techniques for predictive molecular design and analysis. This review delves into the fundamental aspects of chemoinformatics, elucidating the intricate nature of chemical data and the crucial role of molecular descriptors in unveiling the underlying molecular properties. Molecular descriptors, including 2D fingerprints and topological indices, in conjunction with the structure-activity relationships (SARs), are pivotal in unlocking the pathway to small-molecule drug discovery. Technical intricacies of developing robust ML-QSAR models, including feature selection, model validation, and performance evaluation, are discussed herewith. Various ML algorithms, such as regression analysis and support vector machines, are showcased in the text for their ability to predict and comprehend the relationships between molecular structures and biological activities. This review serves as a comprehensive guide for researchers, providing an understanding of the synergy between chemoinformatics, QSAR, and ML. Due to embracing these cutting-edge technologies, predictive molecular analysis holds promise for expediting the discovery of novel therapeutic agents in the pharmaceutical sciences.
Collapse
Affiliation(s)
- Sarfaraz K Niazi
- College of Pharmacy, University of Illinois, Chicago, IL 61820, USA
| | - Zamara Mariam
- Zamara Mariam, School of Interdisciplinary Engineering & Sciences (SINES), National University of Sciences & Technology (NUST), Islamabad 24090, Pakistan
| |
Collapse
|
4
|
Shi C, Nie F, Hu Y, Xu Y, Chen L, Ma X, Luo Q. MedChemLens: An Interactive Visual Tool to Support Direction Selection in Interdisciplinary Experimental Research of Medicinal Chemistry. IEEE TRANSACTIONS ON VISUALIZATION AND COMPUTER GRAPHICS 2023; 29:63-73. [PMID: 36166547 DOI: 10.1109/tvcg.2022.3209434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Interdisciplinary experimental science (e.g., medicinal chemistry) refers to the disciplines that integrate knowledge from different scientific backgrounds and involve experiments in the research process. Deciding "in what direction to proceed" is critical for the success of the research in such disciplines, since the time, money, and resource costs of the subsequent research steps depend largely on this decision. However, such a direction identification task is challenging in that researchers need to integrate information from large-scale, heterogeneous materials from all associated disciplines and summarize the related publications of which the core contributions are often showcased in diverse formats. The task also requires researchers to estimate the feasibility and potential in future experiments in the selected directions. In this work, we selected medicinal chemistry as a case and presented an interactive visual tool, MedChemLens, to assist medicinal chemists in choosing their intended directions of research. This task is also known as drug target (i.e., disease-linked proteins) selection. Given a candidate target name, MedChemLens automatically extracts the molecular features of drug compounds from chemical papers and clinical trial records, organizes them based on the drug structures, and interactively visualizes factors concerning subsequent experiments. We evaluated MedChemLens through a within-subjects study (N=16). Compared with the control condition (i.e., unrestricted online search without using our tool), participants who only used MedChemLens reported faster search, better-informed selections, higher confidence in their selections, and lower cognitive load.
Collapse
|
5
|
Priya S, Tripathi G, Singh DB, Jain P, Kumar A. Machine learning approaches and their applications in drug discovery and design. Chem Biol Drug Des 2022; 100:136-153. [PMID: 35426249 DOI: 10.1111/cbdd.14057] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/30/2022] [Accepted: 04/10/2022] [Indexed: 01/04/2023]
Abstract
This review is focused on several machine learning approaches used in chemoinformatics. Machine learning approaches provide tools and algorithms to improve drug discovery. Many physicochemical properties of drugs like toxicity, absorption, drug-drug interaction, carcinogenesis, and distribution have been effectively modeled by QSAR techniques. Machine learning is a subset of artificial intelligence, and this technique has shown tremendous potential in the field of drug discovery. Techniques discussed in this review are capable of modeling non-linear datasets, as well as big data of increasing depth and complexity. Various machine learning-based approaches are being used for drug target prediction, modeling the structure of drug target, binding site prediction, ligand-based similarity searching, de novo designing of ligands with desired properties, developing scoring functions for molecular docking, building QSAR model for biological activity prediction, and prediction of pharmacokinetic and pharmacodynamic properties of ligands. In recent years, these predictive tools and models have achieved good accuracy. By the use of more related input data, relevant parameters, and appropriate algorithms, the accuracy of these predictions can be further improved.
Collapse
Affiliation(s)
- Sonal Priya
- Department of Chemistry, T. N. B. College, TMBU, Bhagalpur, India
| | - Garima Tripathi
- Department of Chemistry, T. N. B. College, TMBU, Bhagalpur, India
| | - Dev Bukhsh Singh
- Department of Biotechnology, Siddharth University, Siddharth Nagar, India
| | - Priyanka Jain
- National Institute of Plant Genome Research, New Delhi, India
| | - Abhijeet Kumar
- Department of Chemistry, Mahatma Gandhi Central University, Motihari, India
| |
Collapse
|
6
|
Jiang Q, Li M, Li H, Chen L. Entrectinib, a new multi-target inhibitor for cancer therapy. Biomed Pharmacother 2022; 150:112974. [PMID: 35447552 DOI: 10.1016/j.biopha.2022.112974] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/29/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022] Open
Abstract
Clinical practice shows that when single-target drugs treat multi-factor diseases such as tumors, cardiovascular system and endocrine system diseases, it is often difficult to achieve good therapeutic effects, and even serious adverse reactions may occur. Multi-target drugs can simultaneously regulate multiple links of disease, improve efficacy, reduce adverse reactions, and improve drug resistance. They are ideal drugs for treating complex diseases, and therefore have become the main direction of drug development. At present, some multi-target drugs have been successfully used in many major diseases. Entrectinib is an oral small molecule inhibitor that targets TRK, ROS1, and ALK. It is used to treat locally advanced or metastatic solid tumors with NTRK1/2/3, ROS1 and ALK gene fusion mutations. It can pass through the blood-brain barrier and is the only TRK inhibitor clinically proven to be effective against primary and metastatic brain diseases. In 2019, entrectinib was approved by the FDA to treat adult patients with ROS1-positive metastatic non-small cell lung cancer. Case reports showed that continuous administration of entrectinib was effective and tolerable. In this review, we give a brief introduction to TKK, ROS1 and ALK, and on this basis, we give a detailed and comprehensive introduction to the mechanism of action, pharmacokinetics, pharmacodynamics, clinical efficacy, tolerability and drug interactions of entrectinib.
Collapse
Affiliation(s)
- Qinghua Jiang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Mingxue Li
- Wuya College of Innovation, School of Pharmacy, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hua Li
- Wuya College of Innovation, School of Pharmacy, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Lixia Chen
- Wuya College of Innovation, School of Pharmacy, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
7
|
Deep learning for retention time prediction in reversed-phase liquid chromatography. J Chromatogr A 2021; 1664:462792. [PMID: 34999303 DOI: 10.1016/j.chroma.2021.462792] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 01/16/2023]
Abstract
Retention time prediction in high-performance liquid chromatography (HPLC) is the subject of many studies since it can improve the identification of unknown molecules in untargeted profiling using HPLC coupled with high-resolution mass spectrometry. Lots of approaches were developed for retention time prediction in liquid chromatography for a different number of molecules considering various molecular properties and machine learning algorithms. The recently built large retention time data set of standard compounds from the Metabolite and Chemical Entity Database (METLIN) allows researchers to create a model that can be used for retention time prediction of small molecules with wide varieties of structures and physicochemical properties. The ability to predict retention times using the largest data set was studied for different architectures of deep learning models that were trained on molecular fingerprints, and SMILES (string representation of a molecule) represented as one-hot matrices. The best result was achieved with a one-dimensional convolutional neural network (1D CNN) that uses SMILES as an input. The proposed model reached the mean absolute error and the median absolute error equal to 34.7 and 18.7 s, respectively, which outperformed the results previously obtained for this data set. The pre-trained 1D CNN on the METLIN SMRT data set was transferred on five other data sets to evaluate the generalization ability.
Collapse
|
8
|
Kell DB. The Transporter-Mediated Cellular Uptake and Efflux of Pharmaceutical Drugs and Biotechnology Products: How and Why Phospholipid Bilayer Transport Is Negligible in Real Biomembranes. Molecules 2021; 26:5629. [PMID: 34577099 PMCID: PMC8470029 DOI: 10.3390/molecules26185629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Over the years, my colleagues and I have come to realise that the likelihood of pharmaceutical drugs being able to diffuse through whatever unhindered phospholipid bilayer may exist in intact biological membranes in vivo is vanishingly low. This is because (i) most real biomembranes are mostly protein, not lipid, (ii) unlike purely lipid bilayers that can form transient aqueous channels, the high concentrations of proteins serve to stop such activity, (iii) natural evolution long ago selected against transport methods that just let any undesirable products enter a cell, (iv) transporters have now been identified for all kinds of molecules (even water) that were once thought not to require them, (v) many experiments show a massive variation in the uptake of drugs between different cells, tissues, and organisms, that cannot be explained if lipid bilayer transport is significant or if efflux were the only differentiator, and (vi) many experiments that manipulate the expression level of individual transporters as an independent variable demonstrate their role in drug and nutrient uptake (including in cytotoxicity or adverse drug reactions). This makes such transporters valuable both as a means of targeting drugs (not least anti-infectives) to selected cells or tissues and also as drug targets. The same considerations apply to the exploitation of substrate uptake and product efflux transporters in biotechnology. We are also beginning to recognise that transporters are more promiscuous, and antiporter activity is much more widespread, than had been realised, and that such processes are adaptive (i.e., were selected by natural evolution). The purpose of the present review is to summarise the above, and to rehearse and update readers on recent developments. These developments lead us to retain and indeed to strengthen our contention that for transmembrane pharmaceutical drug transport "phospholipid bilayer transport is negligible".
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, UK;
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, 2800 Kgs Lyngby, Denmark
- Mellizyme Biotechnology Ltd., IC1, Liverpool Science Park, Mount Pleasant, Liverpool L3 5TF, UK
| |
Collapse
|
9
|
Lester CC, Yan G. A matched molecular pair (MMP) approach for selecting analogs suitable for structure activity relationship (SAR)-based read across. Regul Toxicol Pharmacol 2021; 124:104966. [PMID: 34044089 DOI: 10.1016/j.yrtph.2021.104966] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/12/2021] [Accepted: 05/19/2021] [Indexed: 11/26/2022]
Abstract
One of the most challenging aspects of SAR-based read across is the identification of structurally similar compounds suitable for use as data sources to cover the safety of a target chemical. Matched molecular pair analysis (MMPA) provides a systematic method for mining experimental data for chemical substitutions that may be interpreted in terms of changes in properties. Here we use the relationships between structural substitutions linking a target chemical with an analog determined to be suitable using the expert-judgment based P&G framework of Wu et al. (2010). The relationships are established by applying MMPA to a database of compounds with safety assessed using SAR-based read across to suitable analogs possessing toxicological data. The analysis revealed that only five categories of substitutions per chemical class (aromatic or aliphatic) were necessary to link all molecular pairs. These data are summarized in a workflow outlining a strategy for searching toxicological databases for potential analogs. This approach provides structural comparisons that are interpretable and sensitive to small differences in the local structure of two compounds that may be linked to suitability for read across in contrast to the use of quantitative similarity measures which show little correlation with analog suitability.
Collapse
Affiliation(s)
- Cathy C Lester
- The Procter & Gamble Company, 8700 Mason Montgomery Rd. Mason, OH, 45040, USA.
| | - Gang Yan
- The Procter & Gamble Company, 8700 Mason Montgomery Rd. Mason, OH, 45040, USA
| |
Collapse
|
10
|
Murray D, Petrey D, Honig B. Integrating 3D structural information into systems biology. J Biol Chem 2021; 296:100562. [PMID: 33744294 PMCID: PMC8095114 DOI: 10.1016/j.jbc.2021.100562] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/18/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
Systems biology is a data-heavy field that focuses on systems-wide depictions of biological phenomena necessarily sacrificing a detailed characterization of individual components. As an example, genome-wide protein interaction networks are widely used in systems biology and continuously extended and refined as new sources of evidence become available. Despite the vast amount of information about individual protein structures and protein complexes that has accumulated in the past 50 years in the Protein Data Bank, the data, computational tools, and language of structural biology are not an integral part of systems biology. However, increasing effort has been devoted to this integration, and the related literature is reviewed here. Relationships between proteins that are detected via structural similarity offer a rich source of information not available from sequence similarity, and homology modeling can be used to leverage Protein Data Bank structures to produce 3D models for a significant fraction of many proteomes. A number of structure-informed genomic and cross-species (i.e., virus–host) interactomes will be described, and the unique information they provide will be illustrated with a number of examples. Tissue- and tumor-specific interactomes have also been developed through computational strategies that exploit patient information and through genetic interactions available from increasingly sensitive screens. Strategies to integrate structural information with these alternate data sources will be described. Finally, efforts to link protein structure space with chemical compound space offer novel sources of information in drug design, off-target identification, and the identification of targets for compounds found to be effective in phenotypic screens.
Collapse
Affiliation(s)
- Diana Murray
- Department of Systems Biology, Columbia University, New York, New York, USA
| | - Donald Petrey
- Department of Systems Biology, Columbia University, New York, New York, USA
| | - Barry Honig
- Department of Systems Biology, Department of Biochemistry and Molecular Biophysics, Department of Medicine, Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, New York, USA.
| |
Collapse
|
11
|
Yang S, Ye Q, Ding J, Yin, Lu A, Chen X, Hou T, Cao D. Current advances in ligand‐based target prediction. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2020. [DOI: 10.1002/wcms.1504] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Su‐Qing Yang
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan China
| | - Qing Ye
- College of Pharmaceutical Sciences Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University Hangzhou, Zhejiang China
| | - Jun‐Jie Ding
- Beijing Institute of Pharmaceutical Chemistry Beijing China
| | - 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 Hunan China
| | - Ai‐Ping Lu
- Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine Hong Kong Baptist University Hong Kong China
| | - Xiang Chen
- 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 Hunan China
| | - Ting‐Jun Hou
- College of Pharmaceutical Sciences Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University Hangzhou, Zhejiang China
| | - Dong‐Sheng Cao
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan China
- Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine Hong Kong Baptist University Hong Kong China
| |
Collapse
|
12
|
Dunkel A, Hofmann T, Di Pizio A. In Silico Investigation of Bitter Hop-Derived Compounds and Their Cognate Bitter Taste Receptors. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:10414-10423. [PMID: 32027492 DOI: 10.1021/acs.jafc.9b07863] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The typical bitter taste of beer is caused by adding hops (Humulus lupulus L.) during the wort boiling process. The bitter taste of hop-derived compounds was found to be mediated by three bitter taste receptors: TAS2R1, TAS2R14, and TAS2R40. In this work, structural bioinformatics analyses were used to characterize the binding modes of trans-isocohumulone, trans-isohumulone, trans-isoadhumulone, cis-isocohumulone, cis-isohumulone, cis-isoadhumulone, cohumulone, humulone, adhumulone, and 8-prenylnaringenin into the orthosteric binding site of their cognate receptors. A conserved asparagine in transmembrane 3 was found to be essential for the recognition of hop-derived compounds, whereas the surrounding residues in the binding site of the three receptors encode the ligand specificity. Hop-derived compounds are renowned bioactive molecules and are considered as potential hit molecules for drug discovery to treat metabolic diseases. A chemoinformatics analysis revealed that hop-derived compounds cluster in a different region of the chemical space compared to known bitter food-derived compounds, pinpointing hop-derived compounds as a very peculiar class of bitter compounds.
Collapse
Affiliation(s)
- Andreas Dunkel
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner Straße 34, D-85354 Freising, Germany
| | - Thomas Hofmann
- Chair of Food Chemistry and Molecular Sensory Science, Technical University of Munich, Lise-Meitner-Straße 34, D-85354 Freising, Germany
| | - Antonella Di Pizio
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner Straße 34, D-85354 Freising, Germany
| |
Collapse
|
13
|
Samanta S, O’Hagan S, Swainston N, Roberts TJ, Kell DB. VAE-Sim: A Novel Molecular Similarity Measure Based on a Variational Autoencoder. Molecules 2020; 25:E3446. [PMID: 32751155 PMCID: PMC7435890 DOI: 10.3390/molecules25153446] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/21/2020] [Accepted: 07/28/2020] [Indexed: 01/13/2023] Open
Abstract
Molecular similarity is an elusive but core "unsupervised" cheminformatics concept, yet different "fingerprint" encodings of molecular structures return very different similarity values, even when using the same similarity metric. Each encoding may be of value when applied to other problems with objective or target functions, implying that a priori none are "better" than the others, nor than encoding-free metrics such as maximum common substructure (MCSS). We here introduce a novel approach to molecular similarity, in the form of a variational autoencoder (VAE). This learns the joint distribution p(z|x) where z is a latent vector and x are the (same) input/output data. It takes the form of a "bowtie"-shaped artificial neural network. In the middle is a "bottleneck layer" or latent vector in which inputs are transformed into, and represented as, a vector of numbers (encoding), with a reverse process (decoding) seeking to return the SMILES string that was the input. We train a VAE on over six million druglike molecules and natural products (including over one million in the final holdout set). The VAE vector distances provide a rapid and novel metric for molecular similarity that is both easily and rapidly calculated. We describe the method and its application to a typical similarity problem in cheminformatics.
Collapse
Affiliation(s)
- Soumitra Samanta
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, UK; (S.S.); (N.S.); (T.J.R.)
| | - Steve O’Hagan
- Department of Chemistry, The Manchester Institute of Biotechnology, The University of Manchester, 131 Princess St, Manchester M1 7DN, UK;
| | - Neil Swainston
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, UK; (S.S.); (N.S.); (T.J.R.)
| | - Timothy J. Roberts
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, UK; (S.S.); (N.S.); (T.J.R.)
| | - Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, UK; (S.S.); (N.S.); (T.J.R.)
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, 2800 Kgs Lyngby, Denmark
| |
Collapse
|
14
|
Martinez-Mayorga K, Madariaga-Mazon A, Medina-Franco JL, Maggiora G. The impact of chemoinformatics on drug discovery in the pharmaceutical industry. Expert Opin Drug Discov 2020; 15:293-306. [PMID: 31965870 DOI: 10.1080/17460441.2020.1696307] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Introduction: Even though there have been substantial advances in our understanding of biological systems, research in drug discovery is only just now beginning to utilize this type of information. The single-target paradigm, which exemplifies the reductionist approach, remains a mainstay of drug research today. A deeper view of the complexity involved in drug discovery is necessary to advance on this field.Areas covered: This perspective provides a summary of research areas where cheminformatics has played a key role in drug discovery, including of the available resources as well as a personal perspective of the challenges still faced in the field.Expert opinion: Although great strides have been made in the handling and analysis of biological and pharmacological data, more must be done to link the data to biological pathways. This is crucial if one is to understand how drugs modify disease phenotypes, although this will involve a shift from the single drug/single target paradigm that remains a mainstay of drug research. Moreover, such a shift would require an increased awareness of the role of physiology in the mechanism of drug action, which will require the introduction of new mathematical, computer, and biological methods for chemoinformaticians to be trained in.
Collapse
Affiliation(s)
| | | | - José L Medina-Franco
- Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | |
Collapse
|
15
|
Gagic Z, Ruzic D, Djokovic N, Djikic T, Nikolic K. In silico Methods for Design of Kinase Inhibitors as Anticancer Drugs. Front Chem 2020; 7:873. [PMID: 31970149 PMCID: PMC6960140 DOI: 10.3389/fchem.2019.00873] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
Rational drug design implies usage of molecular modeling techniques such as pharmacophore modeling, molecular dynamics, virtual screening, and molecular docking to explain the activity of biomolecules, define molecular determinants for interaction with the drug target, and design more efficient drug candidates. Kinases play an essential role in cell function and therefore are extensively studied targets in drug design and discovery. Kinase inhibitors are clinically very important and widely used antineoplastic drugs. In this review, computational methods used in rational drug design of kinase inhibitors are discussed and compared, considering some representative case studies.
Collapse
Affiliation(s)
- Zarko Gagic
- Department of Pharmaceutical Chemistry, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Nemanja Djokovic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Teodora Djikic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
16
|
When global and local molecular descriptors are more than the sum of its parts: Simple, But Not Simpler? Mol Divers 2019; 24:913-932. [PMID: 31659696 DOI: 10.1007/s11030-019-10002-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/09/2019] [Indexed: 01/29/2023]
Abstract
In this report, we introduce a set of aggregation operators (AOs) to calculate global and local (group and atom type) molecular descriptors (MDs) as a generalization of the classical approach of molecular encoding using the sum of the atomic (or fragment) contributions. These AOs are implemented in a new and free software denominated MD-LOVIs ( http://tomocomd.com/md-lovis ), which allows for the calculation of MDs from atomic weights vector and LOVIs (local vertex invariants). This software was developed in Java programming language and employed the Chemical Development Kit (CDK) library for handling chemical structures and the calculation of atomic weights. An analysis of the complexities of the algorithms presented herein demonstrates that these aspects were efficiently implemented. The calculation speed experiments show that the MD-LOVIs software has satisfactory behavior when compared to software such as Padel, CDKDescriptor, DRAGON and Bluecal software. Shannon's entropy (SE)-based variability studies demonstrate that MD-LOVIs yields indices with greater information content when compared to those of popular academic and commercial software. A principal component analysis reveals that our approach captures chemical information orthogonal to that codified by the DRAGON, Padel and Mold2 software, as a result of the several generalizations in MD-LOVIs not used in other programs. Lastly, three QSARs were built using multiple linear regression with genetic algorithms, and the statistical parameters of these models demonstrate that the MD-LOVIs indices obtained with AOs yield better performance than those obtained when the summation operator is used exclusively. Moreover, it is also revealed that the MD-LOVIs indices yield models with comparable to superior performance when compared to other QSAR methodologies reported in the literature, despite their simplicity. The studies performed herein collectively demonstrated that MD-LOVIs software generates indices as simple as possible, but not simpler and that use of AOs enhances the diversity of the chemical information codified, which consequently improves the performance of traditional MDs.
Collapse
|
17
|
Kausar S, Falcao AO. A visual approach for analysis and inference of molecular activity spaces. J Cheminform 2019; 11:63. [PMID: 33430986 PMCID: PMC6805449 DOI: 10.1186/s13321-019-0386-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/05/2019] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Molecular space visualization can help to explore the diversity of large heterogeneous chemical data, which ultimately may increase the understanding of structure-activity relationships (SAR) in drug discovery projects. Visual SAR analysis can therefore be useful for library design, chemical classification for their biological evaluation and virtual screening for the selection of compounds for synthesis or in vitro testing. As such, computational approaches for molecular space visualization have become an important issue in cheminformatics research. The proposed approach uses molecular similarity as the sole input for computing a probabilistic surface of molecular activity (PSMA). This similarity matrix is transformed in 2D using different dimension reduction algorithms (Principal Coordinates Analysis ( PCooA), Kruskal multidimensional scaling, Sammon mapping and t-SNE). From this projection, a kernel density function is applied to compute the probability of activity for each coordinate in the new projected space. RESULTS This methodology was tested over four different quantitative structure-activity relationship (QSAR) binary classification data sets and the PSMAs were computed for each. The generated maps showed internal consistency with active molecules grouped together for all data sets and all dimensionality reduction algorithms. To validate the quality of the generated maps, the 2D coordinates of test molecules were computed into the new reference space using a data transformation matrix. In total sixteen PSMAs were built, and their performance was assessed using the Area Under Curve (AUC) and the Matthews Coefficient Correlation (MCC). For the best projections for each data set, AUC testing results ranged from 0.87 to 0.98 and the MCC scores ranged from 0.33 to 0.77, suggesting this methodology can validly capture the complexities of the molecular activity space. All four mapping functions provided generally good results yet the overall performance of PCooA and t-SNE was slightly better than Sammon mapping and Kruskal multidimensional scaling. CONCLUSIONS Our result showed that by using an appropriate combination of metric space representation and dimensionality reduction applied over metric spaces it is possible to produce a visual PSMA for which its consistency has been validated by using this map as a classification model. The produced maps can be used as prediction tools as it is simple to project any molecule into this new reference space as long as the similarities to the molecules used to compute the initial similarity matrix can be computed.
Collapse
Affiliation(s)
- Samina Kausar
- LaSIGE, Departamento de Informática, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- BioISI: Biosystems & Integrative Sciences Institute, Faculdade de Ciencias, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Andre O. Falcao
- LaSIGE, Departamento de Informática, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- BioISI: Biosystems & Integrative Sciences Institute, Faculdade de Ciencias, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| |
Collapse
|
18
|
Wang Y, Yella J, Jegga AG. Transcriptomic Data Mining and Repurposing for Computational Drug Discovery. Methods Mol Biol 2019; 1903:73-95. [PMID: 30547437 DOI: 10.1007/978-1-4939-8955-3_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Conventional drug discovery in general is costly and time-consuming with extremely low success and relatively high attrition rates. The disparity between high cost of drug discovery and vast unmet medical needs resulted in advent of an increasing number of computational approaches that can "connect" disease with a candidate therapeutic. This includes computational drug repurposing or repositioning wherein the goal is to discover a new indication for an approved drug. Computational drug discovery approaches that are commonly used are similarity-based wherein network analysis or machine learning-based methods are used. One such approach is matching gene expression signatures from disease to those from small molecules, commonly referred to as connectivity mapping. In this chapter, we will focus on how publicly available existing transcriptomic data from diseases can be reused to identify novel candidate therapeutics and drug repositioning candidates. To elucidate these, we will present two case studies: (1) using transcriptional signature similarity or positive correlation to identify novel small molecules that are similar to an approved drug and (2) identifying candidate therapeutics via reciprocal connectivity or negative correlation between transcriptional signatures from a disease and small molecule.
Collapse
Affiliation(s)
- Yunguan Wang
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jaswanth Yella
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Computer Science, University of Cincinnati College of Engineering, Cincinnati, OH, USA
| | - Anil G Jegga
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA. .,Department of Computer Science, University of Cincinnati College of Engineering, Cincinnati, OH, USA.
| |
Collapse
|
19
|
García-Jacas CR, Cabrera-Leyva L, Marrero-Ponce Y, Suárez-Lezcano J, Cortés-Guzmán F, Pupo-Meriño M, Vivas-Reyes R. Choquet integral-based fuzzy molecular characterizations: when global definitions are computed from the dependency among atom/bond contributions (LOVIs/LOEIs). J Cheminform 2018; 10:51. [PMID: 30362050 PMCID: PMC6755596 DOI: 10.1186/s13321-018-0306-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/15/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Several topological (2D) and geometric (3D) molecular descriptors (MDs) are calculated from local vertex/edge invariants (LOVIs/LOEIs) by performing an aggregation process. To this end, norm-, mean- and statistic-based (non-fuzzy) operators are used, under the assumption that LOVIs/LOEIs are independent (orthogonal) values of one another. These operators are based on additive and/or linear measures and, consequently, they cannot be used to encode information from interrelated criteria. Thus, as LOVIs/LOEIs are not orthogonal values, then non-additive (fuzzy) measures can be used to encode the interrelation among them. RESULTS General approaches to compute fuzzy 2D/3D-MDs from the contribution of each atom (LOVIs) or covalent bond (LOEIs) within a molecule are proposed, by using the Choquet integral as fuzzy aggregation operator. The Choquet integral-based operator is rather different from the other operators often used for the 2D/3D-MDs calculation. It performs a reordering step to fuse the LOVIs/LOEIs according to their magnitudes and, in addition, it considers the interrelation among them through a fuzzy measure. With this operator, fuzzy definitions can be derived from traditional or recent MDs; for instance, fuzzy Randic-like connectivity indices, fuzzy Balaban-like indices, fuzzy Kier-Hall connectivity indices, among others. To demonstrate the feasibility of using this operator, the QuBiLS-MIDAS 3D-MDs were used as study case and, as a result, a module was built into the corresponding software to compute them ( http://tomocomd.com/qubils-midas ). Thus, it is the only software reported in the literature that can be employed to determine Choquet integral-based fuzzy MDs. Moreover, regression models were created on eight chemical datasets. In this way, a comparison between the results achieved by the models based on the non-fuzzy QuBiLS-MIDAS 3D-MDs with regard to the ones achieved by the models based on the fuzzy QuBiLS-MIDAS 3D-MDs was made. As a result, the models built with the fuzzy QuBiLS-MIDAS 3D-MDs achieved the best performance, which was statistically corroborated through the Wilcoxon signed-rank test. CONCLUSIONS All in all, it can be concluded that the Choquet integral constitutes a prominent alternative to compute fuzzy 2D/3D-MDs from LOVIs/LOEIs. In this way, better characterizations of the compounds can be obtained, which will be ultimately useful in enhancing the modelling ability of existing traditional 2D/3D-MDs.
Collapse
Affiliation(s)
- César R. García-Jacas
- Instituto de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Lisset Cabrera-Leyva
- Grupo de Investigación de Inteligencia Artificial (AIRES), Facultad de Informática, Universidad de Camagüey, Camagüey, Cuba
| | - Yovani Marrero-Ponce
- Grupo de Medicina Molecular y Traslacional (MeM&T), Colegio de Ciencias de la Salud (COCSA), Escuela de Medicina, Edificio de Especialidades Médicas, Universidad San Francisco de Quito (USFQ), Quito, Pichincha Ecuador
- Grupo de Investigación Ambiental (GIA), Programas Ambientales, Facultad de Ingenierías, Fundacion Universitaria Tecnologico Comfenalco – Cartagena, Cr 44 DN 30 A, 91, Cartagena, Bolívar Colombia
| | - José Suárez-Lezcano
- Pontificia Universidad Católica del Ecuador Sede Esmeraldas (PUCESE), Esmeraldas, Ecuador
| | - Fernando Cortés-Guzmán
- Instituto de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Mario Pupo-Meriño
- Grupo de Investigación de Bioinformática, Universidad de las Ciencias Informáticas (UCI), La Habana, Cuba
| | - Ricardo Vivas-Reyes
- Grupo de Química Cuántica y Teórica, Facultad de Ciencias Exactas y Naturales, Programa de Química, Universidad de Cartagena, Campus de San Pablo, Cartagena, Colombia
- Grupo CipTec, Facultad de Ingenierias, Fundacion Universitaria Tecnologico Comfenalco – Cartagena, Cr 44 DN 30 A, 91, Cartagena, Bolívar Colombia
| |
Collapse
|
20
|
Lo YC, Rensi SE, Torng W, Altman RB. Machine learning in chemoinformatics and drug discovery. Drug Discov Today 2018; 23:1538-1546. [PMID: 29750902 DOI: 10.1016/j.drudis.2018.05.010] [Citation(s) in RCA: 451] [Impact Index Per Article: 75.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/29/2018] [Accepted: 05/02/2018] [Indexed: 01/03/2023]
Abstract
Chemoinformatics is an established discipline focusing on extracting, processing and extrapolating meaningful data from chemical structures. With the rapid explosion of chemical 'big' data from HTS and combinatorial synthesis, machine learning has become an indispensable tool for drug designers to mine chemical information from large compound databases to design drugs with important biological properties. To process the chemical data, we first reviewed multiple processing layers in the chemoinformatics pipeline followed by the introduction of commonly used machine learning models in drug discovery and QSAR analysis. Here, we present basic principles and recent case studies to demonstrate the utility of machine learning techniques in chemoinformatics analyses; and we discuss limitations and future directions to guide further development in this evolving field.
Collapse
Affiliation(s)
- Yu-Chen Lo
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Stefano E Rensi
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Wen Torng
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Russ B Altman
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|