1
|
Petrov K, Bender A. An Open-Source Implementation of the Scaffold Identification and Naming System (SCINS) and Example Applications. J Chem Inf Model 2024; 64:7905-7916. [PMID: 39404472 PMCID: PMC11523071 DOI: 10.1021/acs.jcim.4c01314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024]
Abstract
Organizing and partitioning sets of chemical structures is of considerable practical significance, e.g., in compound library analysis and the postprocessing of screening hit lists. Approaches such as unsupervised clustering are computationally demanding and dataset-dependent; on the other hand, rule-based methods, such as those based on Murcko scaffolds, have linear time complexity but are often too fine-grained, leading to a large number of singletons or sparsely populated classes. An alternative rule-based method that seeks to achieve an optimal balance when grouping compounds into sets is the 'Scaffold Identification and Naming System' (SCINS). To facilitate public use of this previously published method, here, we provide an open-source Python implementation of SCINS, dependent only on RDKit. We show that SCINS can be useful in identifying sparsely and densely populated regions in chemical space in large databases, here exemplified with Enamine REAL Diverse and ChEMBL. We find that Enamine REAL Diverse covers a much smaller SCINS space relative to ChEMBL, whereas the opposite is true when Murcko and generic Murcko scaffolds are considered. Additionally, we show that SCINS can result in chemically intuitive grouping of medium-sized sets of bioactive compounds, which can be useful in compound selection from virtual screening campaigns as well as postprocessing of experimental hit lists. Hence, in this work, we provide both an open-source implementation of SCINS and its characterization with relevant use cases.
Collapse
Affiliation(s)
- Kamen
P. Petrov
- Pangea
Bio, Pangea Botanica GmbH, Hardenbergstrasse 32, 10623 Berlin, Germany
| | - Andreas Bender
- Pangea
Bio, Pangea Botanica GmbH, Hardenbergstrasse 32, 10623 Berlin, Germany
- Centre
for Molecular Informatics, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Rd, CB2
1EW Cambridge, United
Kingdom
| |
Collapse
|
2
|
Avellaneda-Tamayo JF, Agudo-Muñoz NA, Sánchez-Galán JE, López-Pérez JL, Medina-Franco JL. Chemoinformatic Characterization of NAPROC-13: A Database for Natural Product 13C NMR Dereplication. JOURNAL OF NATURAL PRODUCTS 2024; 87:2216-2229. [PMID: 39269718 PMCID: PMC11443490 DOI: 10.1021/acs.jnatprod.4c00530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
Natural products (NPs) are secondary metabolites of natural origin with broad applications across various human activities, particularly the discovery of bioactive compounds. Structural elucidation of new NPs entails significant cost and effort. On the other hand, the dereplication of known compounds is crucial for the early exclusion of irrelevant compounds in contemporary pharmaceutical research. NAPROC-13 stands out as a publicly accessible database, providing structural and 13C NMR spectroscopic information for over 25 000 compounds, rendering it a pivotal resource in natural product (NP) research, favoring open science. This study seeks to quantitatively analyze the chemical content, structural diversity, and chemical space coverage of NPs within NAPROC-13, compared to FDA-approved drugs and a very diverse subset of NPs, UNPD-A. Findings indicated that NPs in NAPROC-13 exhibit properties comparable to those in UNPD-A, albeit showcasing a notably diverse array of structural content, scaffolds, ring systems of pharmaceutical interest, and molecular fragments. NAPROC-13 covers a specific region of the chemical multiverse (a generalization of the chemical space from different chemical representations) regarding physicochemical properties and a region as broad as UNPD-A in terms of the structural features represented by fingerprints.
Collapse
Affiliation(s)
- Juan F. Avellaneda-Tamayo
- DIFACQUIM
Research Group, Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Mexico City 04510, Mexico
| | - Naicolette A. Agudo-Muñoz
- Science
and Technology Faculty, Universidad Tecnológica de Panamá,
Campus Metropolitano Víctor Levi Sasso, Avenida Universidad Tecnológica, Vía Puente Centenario, Panama City 0819-07289, Panama
- Grupo
de Investigación en Biotecnología, Bioinformática
y Biología de Sistemas (GIBBS), Universidad Tecnológica
de Panama, Panama City, Panama
| | - Javier E. Sánchez-Galán
- Facultad
de Ingeniería de Sistemas Computacionales, Universidad Tecnológica
de Panamá, Campus Metropolitano Víctor Levi Sasso, Avenida Universidad Tecnológica, Vía
Puente Centenario, Panama City 0819-07289, Panama
- Grupo
de Investigación en Biotecnología, Bioinformática
y Biología de Sistemas (GIBBS), Universidad Tecnológica
de Panama, Panama City, Panama
| | - José L. López-Pérez
- Departamento
de Ciencias Farmacéuticas, Área de Química Farmacéutica,
Facultad de Farmacia, CIETUS, IBSAL, Campus Miguel de Unamuno, University of Salamanca, 37007, Salamanca, Spain
- Departamento
de Farmacología, Facultad de Medicina, CIPFAR, Universidad de Panamá, Panama City, Panama
| | - José L. Medina-Franco
- DIFACQUIM
Research Group, Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Mexico City 04510, Mexico
| |
Collapse
|
3
|
Zhao Y, Zhang Z, Kong X, Wang K, Wang Y, Jia J, Li H, Tian S. Prediction of Drug-Induced Liver Injury: From Molecular Physicochemical Properties and Scaffold Architectures to Machine Learning Approaches. Chem Biol Drug Des 2024; 104:e14607. [PMID: 39179521 DOI: 10.1111/cbdd.14607] [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: 05/06/2024] [Revised: 07/24/2024] [Accepted: 08/01/2024] [Indexed: 08/26/2024]
Abstract
The process of developing new drugs is widely acknowledged as being time-intensive and requiring substantial financial investment. Despite ongoing efforts to reduce time and expenses in drug development, ensuring medication safety remains an urgent problem. One of the major problems involved in drug development is hepatotoxicity, specifically known as drug-induced liver injury (DILI). The popularity of new drugs often poses a significant barrier during development and frequently leads to their recall after launch. In silico methods have many advantages compared with traditional in vivo and in vitro assays. To establish a more precise and reliable prediction model, it is necessary to utilize an extensive and high-quality database consisting of information on drug molecule properties and structural patterns. In addition, we should also carefully select appropriate molecular descriptors that can be used to accurately depict compound characteristics. The aim of this study was to conduct a comprehensive investigation into the prediction of DILI. First, we conducted a comparative analysis of the physicochemical properties of extensively well-prepared DILI-positive and DILI-negative compounds. Then, we used classic substructure dissection methods to identify structural pattern differences between these two different types of chemical molecules. These findings indicate that it is not feasible to establish property or substructure-based rules for distinguishing between DILI-positive and DILI-negative compounds. Finally, we developed quantitative classification models for predicting DILI using the naïve Bayes classifier (NBC) and recursive partitioning (RP) machine learning techniques. The optimal DILI prediction model was obtained using NBC, which combines 21 physicochemical properties, the VolSurf descriptors and the LCFP_10 fingerprint set. This model achieved a global accuracy (GA) of 0.855 and an area under the curve (AUC) of 0.704 for the training set, while the corresponding values were 0.619 and 0.674 for the test set, respectively. Moreover, indicative substructural fragments favorable or unfavorable for DILI were identified from the best naïve Bayesian classification model. These findings may help prioritize lead compounds in the early stage of drug development pipelines.
Collapse
Affiliation(s)
- Yulong Zhao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zhoudong Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xiaotian Kong
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| | - Kai Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yaxuan Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jie Jia
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Huanqiu Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Sheng Tian
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| |
Collapse
|
4
|
Heinzke AL, Pahl A, Zdrazil B, Leach AR, Waldmann H, Young RJ, Leeson PD. Occurrence of "Natural Selection" in Successful Small Molecule Drug Discovery. J Med Chem 2024; 67:11226-11241. [PMID: 38949112 PMCID: PMC11247505 DOI: 10.1021/acs.jmedchem.4c00811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/08/2024] [Accepted: 06/13/2024] [Indexed: 07/02/2024]
Abstract
Published compounds from ChEMBL version 32 are used to seek evidence for the occurrence of "natural selection" in drug discovery. Three measures of natural product (NP) character were applied, to compare time- and target-matched compounds reaching the clinic (clinical compounds in phase 1-3 development and approved drugs) with background compounds (reference compounds). Pseudo-NPs (PNPs), containing NP fragments combined in ways inaccessible by nature, are increasing over time, reaching 67% of clinical compounds first disclosed since 2010. PNPs are 54% more likely to be found in post-2008 clinical versus reference compounds. The majority of target classes show increased clinical compound NP character versus their reference compounds. Only 176 NP fragments appear in >1000 clinical compounds published since 2008, yet these make up on average 63% of the clinical compound's core scaffolds. There is untapped potential awaiting exploitation, by applying nature's building blocks─"natural intelligence"─to drug design.
Collapse
Affiliation(s)
- A. Lina Heinzke
- European
Molecular Biology Laboratory, European Bioinformatics
Institute, Wellcome Genome Campus, Hinxton CB10 1SD, Cambridgeshire, U.K.
| | - Axel Pahl
- Compound
Management and Screening Center, Max-Planck-Institute
of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Barbara Zdrazil
- European
Molecular Biology Laboratory, European Bioinformatics
Institute, Wellcome Genome Campus, Hinxton CB10 1SD, Cambridgeshire, U.K.
| | - Andrew R. Leach
- European
Molecular Biology Laboratory, European Bioinformatics
Institute, Wellcome Genome Campus, Hinxton CB10 1SD, Cambridgeshire, U.K.
| | - Herbert Waldmann
- Department
of Chemical Biology, Max-Planck-Institute
of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
- Faculty
of Chemistry and Chemical Biology, Technical
University Dortmund, Otto-Hahn-Straße 6, 44227 Dortmund, Germany
| | | | - Paul D. Leeson
- Paul Leeson
Consulting Ltd., Nuneaton CV13 6LZ, Warwickshire, U.K.
| |
Collapse
|
5
|
Snyder SH, Vignaux PA, Ozalp MK, Gerlach J, Puhl AC, Lane TR, Corbett J, Urbina F, Ekins S. The Goldilocks paradigm: comparing classical machine learning, large language models, and few-shot learning for drug discovery applications. Commun Chem 2024; 7:134. [PMID: 38866916 PMCID: PMC11169557 DOI: 10.1038/s42004-024-01220-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
Recent advances in machine learning (ML) have led to newer model architectures including transformers (large language models, LLMs) showing state of the art results in text generation and image analysis as well as few-shot learning (FSLC) models which offer predictive power with extremely small datasets. These new architectures may offer promise, yet the 'no-free lunch' theorem suggests that no single model algorithm can outperform at all possible tasks. Here, we explore the capabilities of classical (SVR), FSLC, and transformer models (MolBART) over a range of dataset tasks and show a 'goldilocks zone' for each model type, in which dataset size and feature distribution (i.e. dataset "diversity") determines the optimal algorithm strategy. When datasets are small ( < 50 molecules), FSLC tend to outperform both classical ML and transformers. When datasets are small-to-medium sized (50-240 molecules) and diverse, transformers outperform both classical models and few-shot learning. Finally, when datasets are of larger and of sufficient size, classical models then perform the best, suggesting that the optimal model to choose likely depends on the dataset available, its size and diversity. These findings may help to answer the perennial question of which ML algorithm is to be used when faced with a new dataset.
Collapse
Affiliation(s)
- Scott H Snyder
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Patricia A Vignaux
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Mustafa Kemal Ozalp
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Jacob Gerlach
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Ana C Puhl
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Thomas R Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - John Corbett
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Fabio Urbina
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA.
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA.
| |
Collapse
|
6
|
Gadiya Y, Shetty S, Hofmann-Apitius M, Gribbon P, Zaliani A. Exploring SureChEMBL from a drug discovery perspective. Sci Data 2024; 11:507. [PMID: 38755219 PMCID: PMC11099139 DOI: 10.1038/s41597-024-03371-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024] Open
Abstract
In the pharmaceutical industry, the patent protection of drugs and medicines is accorded importance because of the high costs involved in the development of novel drugs. Over the years, researchers have analyzed patent documents to identify freedom-to-operate spaces for novel drug candidates. To assist this, several well-established public patent document data repositories have enabled automated methodologies for extracting information on therapeutic agents. In this study, we delve into one such publicly available patent database, SureChEMBL, which catalogues patent documents related to life sciences. Our exploration begins by identifying patent compounds across public chemical data resources, followed by pinpointing sections in patent documents where the chemical annotations were found. Next, we exhibit the potential of compounds to serve as drug candidates by evaluating their conformity to drug-likeness criteria. Lastly, we examine the drug development stage reported for these compounds to understand their clinical success. In summary, our investigation aims at providing a comprehensive overview of the patent compounds catalogued in SureChEMBL, assessing their relevance to pharmaceutical drug discovery.
Collapse
Affiliation(s)
- Yojana Gadiya
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525, Hamburg, Germany.
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Theodor Stern Kai 7, 60590, Frankfurt, Germany.
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany.
| | - Simran Shetty
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525, Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Theodor Stern Kai 7, 60590, Frankfurt, Germany
- Hamburg University of Applied Sciences (HAW), 20099, Hamburg, Germany
| | - Martin Hofmann-Apitius
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53757, Sankt Augustin, Germany
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525, Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Theodor Stern Kai 7, 60590, Frankfurt, Germany
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525, Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Theodor Stern Kai 7, 60590, Frankfurt, Germany
| |
Collapse
|
7
|
Tang SA, Fults A, Boyd SR, Gattu N, Tran KA, Fan J, MacKenzie KR, Palzkill T, Young DW, Chamakuri S. Expanding Complex Morpholines Using Systematic Chemical Diversity. Org Lett 2024; 26:3493-3497. [PMID: 38506470 DOI: 10.1021/acs.orglett.4c00528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
The morpholine heterocycle is a structural unit found in many bioactive compounds and FDA-approved drugs, but the generation of more complex C-functionalized morpholine derivatives remains considerably underexplored. Using systematic chemical diversity (SCD), a concept that guides the expansion of saturated drug-like scaffolds through regiochemical and stereochemical variation, we describe the synthesis of a collection of methyl-substituted morpholine acetic acid esters starting from enantiomerically pure amino acids and amino alcohols. In total, 24 diverse substituted morpholines were produced that vary systematically in regiochemistry and stereochemistry (relative and absolute). These diverse C-substituted morpholines can be directly applied in fragment screening or incorporated as building blocks in medicinal chemistry and library synthesis.
Collapse
Affiliation(s)
- Sunny Ann Tang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Afton Fults
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Shelton R Boyd
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Nikhil Gattu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Kevin A Tran
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Jiayi Fan
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Kevin R MacKenzie
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Timothy Palzkill
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Damian W Young
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Srinivas Chamakuri
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| |
Collapse
|
8
|
Avellaneda-Tamayo JF, Chávez-Hernández AL, Prado-Romero DL, Medina-Franco JL. Chemical Multiverse and Diversity of Food Chemicals. J Chem Inf Model 2024; 64:1229-1244. [PMID: 38356237 PMCID: PMC10900296 DOI: 10.1021/acs.jcim.3c01617] [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: 10/05/2023] [Revised: 02/03/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
Food chemicals have a fundamental role in our lives, with an extended impact on nutrition, disease prevention, and marked economic implications in the food industry. The number of food chemical compounds in public databases has substantially increased in the past few years, which can be characterized using chemoinformatics approaches. We and other groups explored public food chemical libraries containing up to 26,500 compounds. This study aimed to analyze the chemical contents, diversity, and coverage in the chemical space of food chemicals and additives and, from here on, food components. The approach to food components addressed in this study is a public database with more than 70,000 compounds, including those predicted via omics techniques. It was concluded that food components have distinctive physicochemical properties and constitutional descriptors despite sharing many chemical structures with natural products. Food components, on average, have large molecular weights and several apolar structures with saturated hydrocarbons. Compared to reference databases, food component structures have low scaffold and fingerprint-based diversity and high structural complexity, as measured by the fraction of sp3 carbons. These structural features are associated with a large fraction of macronutrients as lipids. Lipids in food components were decompiled by an analysis of the maximum common substructures. The chemical multiverse representation of food chemicals showed a larger coverage of chemical space than natural products and FDA-approved drugs by using different sets of representations.
Collapse
Affiliation(s)
- Juan F. Avellaneda-Tamayo
- DIFACQUIM Research Group, Department
of Pharmacy, School of Chemistry, Universidad
Nacional Autónoma de México, Avenida Universidad 3000, Mexico City 04510, Mexico
| | - Ana L. Chávez-Hernández
- DIFACQUIM Research Group, Department
of Pharmacy, School of Chemistry, Universidad
Nacional Autónoma de México, Avenida Universidad 3000, Mexico City 04510, Mexico
| | - Diana L. Prado-Romero
- DIFACQUIM Research Group, Department
of Pharmacy, School of Chemistry, Universidad
Nacional Autónoma de México, Avenida Universidad 3000, Mexico City 04510, Mexico
| | - José L. Medina-Franco
- DIFACQUIM Research Group, Department
of Pharmacy, School of Chemistry, Universidad
Nacional Autónoma de México, Avenida Universidad 3000, Mexico City 04510, Mexico
| |
Collapse
|
9
|
Nitulescu GM. Techniques and Strategies in Drug Design and Discovery. Int J Mol Sci 2024; 25:1364. [PMID: 38338643 PMCID: PMC10855429 DOI: 10.3390/ijms25031364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 01/18/2024] [Indexed: 02/12/2024] Open
Abstract
The process of drug discovery constitutes a highly intricate and formidable undertaking, encompassing the identification and advancement of novel therapeutic entities [...].
Collapse
Affiliation(s)
- George Mihai Nitulescu
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| |
Collapse
|
10
|
Bitencourt-Ferreira G, Villarreal MA, Quiroga R, Biziukova N, Poroikov V, Tarasova O, de Azevedo Junior WF. Exploring Scoring Function Space: Developing Computational Models for Drug Discovery. Curr Med Chem 2024; 31:2361-2377. [PMID: 36944627 DOI: 10.2174/0929867330666230321103731] [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: 06/23/2022] [Revised: 12/15/2022] [Accepted: 12/29/2022] [Indexed: 03/23/2023]
Abstract
BACKGROUND The idea of scoring function space established a systems-level approach to address the development of models to predict the affinity of drug molecules by those interested in drug discovery. OBJECTIVE Our goal here is to review the concept of scoring function space and how to explore it to develop machine learning models to address protein-ligand binding affinity. METHODS We searched the articles available in PubMed related to the scoring function space. We also utilized crystallographic structures found in the protein data bank (PDB) to represent the protein space. RESULTS The application of systems-level approaches to address receptor-drug interactions allows us to have a holistic view of the process of drug discovery. The scoring function space adds flexibility to the process since it makes it possible to see drug discovery as a relationship involving mathematical spaces. CONCLUSION The application of the concept of scoring function space has provided us with an integrated view of drug discovery methods. This concept is useful during drug discovery, where we see the process as a computational search of the scoring function space to find an adequate model to predict receptor-drug binding affinity.
Collapse
Affiliation(s)
| | - Marcos A Villarreal
- CONICET-Departamento de Matemática y Física, Instituto de Investigaciones en Fisicoquímica de Córdoba (INFIQC), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, Córdoba, Argentina
| | - Rodrigo Quiroga
- CONICET-Departamento de Matemática y Física, Instituto de Investigaciones en Fisicoquímica de Córdoba (INFIQC), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, Córdoba, Argentina
| | - Nadezhda Biziukova
- Institute of Biomedical Chemistry, Pogodinskaya Str., 10/8, Moscow, 119121, Russia
| | - Vladimir Poroikov
- Institute of Biomedical Chemistry, Pogodinskaya Str., 10/8, Moscow, 119121, Russia
| | - Olga Tarasova
- Institute of Biomedical Chemistry, Pogodinskaya Str., 10/8, Moscow, 119121, Russia
| | - Walter F de Azevedo Junior
- Pontifical Catholic University of Rio Grande do Sul - PUCRS, Porto Alegre-RS, Brazil
- Specialization Program in Bioinformatics, The Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681 Porto Alegre / RS 90619-900, Brazil
| |
Collapse
|
11
|
Horgan MJ, Zell L, Siewert B, Stuppner H, Schuster D, Temml V. Identification of Novel β-Tubulin Inhibitors Using a Combined In Silico/ In Vitro Approach. J Chem Inf Model 2023; 63:6396-6411. [PMID: 37774242 PMCID: PMC10598795 DOI: 10.1021/acs.jcim.3c00939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Indexed: 10/01/2023]
Abstract
Due to their potential as leads for various therapeutic applications, including as antimitotic and antiparasitic agents, the development of tubulin inhibitors offers promise for drug discovery. In this study, an in silico pharmacophore-based virtual screening approach targeting the colchicine binding site of β-tubulin was employed. Several structure- and ligand-based models for known tubulin inhibitors were generated. Compound databases were virtually screened against the models, and prioritized hits from the SPECS compound library were tested in an in vitro tubulin polymerization inhibition assay for their experimental validation. Out of the 41 SPECS compounds tested, 11 were active tubulin polymerization inhibitors, leading to a prospective true positive hit rate of 26.8%. Two novel inhibitors displayed IC50 values in the range of colchicine. The most potent of which was a novel acetamide-bridged benzodiazepine/benzimidazole derivative with an IC50 = 2.9 μM. The screening workflow led to the identification of diverse inhibitors active at the tubulin colchicine binding site. Thus, the pharmacophore models show promise as valuable tools for the discovery of compounds and as potential leads for the development of cancer therapeutic agents.
Collapse
Affiliation(s)
- Mark James Horgan
- Institute
of Pharmacy/Pharmacognosy, Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Lukas Zell
- Institute
of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria
| | - Bianka Siewert
- Institute
of Pharmacy/Pharmacognosy, Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Hermann Stuppner
- Institute
of Pharmacy/Pharmacognosy, Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Daniela Schuster
- Institute
of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria
| | - Veronika Temml
- Institute
of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria
| |
Collapse
|
12
|
Kashyap K, Mahapatra PP, Ahmed S, Buyukbingol E, Siddiqi MI. Identification of Potential Aldose Reductase Inhibitors Using Convolutional Neural Network-Based in Silico Screening. J Chem Inf Model 2023; 63:6261-6282. [PMID: 37788831 DOI: 10.1021/acs.jcim.3c00547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Aldose reductase (ALR2) is a notable enzyme of the polyol pathway responsible for aggravating diabetic neuropathy complications. The first step begins when it catalyzes the reduction of glucose to sorbitol with NADPH as a coenzyme. Elevated concentrations of sorbitol damage the tissues, leading to complications like neuropathy. Though considerable effort has been pushed toward the successful discovery of potent inhibitors, its discovery still remains an elusive task. To this end, we present a 3D convolutional neural network (3D-CNN) based ALR2 inhibitor classification technique by dealing with snapshots of images captured from 3D chemical structures with multiple rotations as input data. The CNN-based architecture was trained on the 360 sets of image data along each axis and further prediction on the Maybridge library by each of the models. Subjecting the retrieved hits to molecular docking leads to the identification of the top 10 molecules with high binding affinity. The hits displayed a better blood-brain barrier penetration (BBB) score (90% with more than four scores) as compared to standard inhibitors (38%), reflecting the superior BBB penetrating efficiency of the hits. Followed by molecular docking, the biological evaluation spotlighted five compounds as promising ALR2 inhibitors and can be considered as a likely prospect for further structural optimization with medicinal chemistry efforts to improve their inhibition efficacy and consolidate them as new ALR2 antagonists in the future. In addition, the study also demonstrated the usefulness of scaffold analysis of the molecules as a method for investigating the significance of structurally diverse compounds in data-driven studies. For reproducibility and accessibility purposes, all of the source codes used in our study are publicly available.
Collapse
Affiliation(s)
- Kushagra Kashyap
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pinaki Prasad Mahapatra
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Shakil Ahmed
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Erdem Buyukbingol
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, 06100 Ankara, Turkey
| | - Mohammad Imran Siddiqi
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
13
|
Di Lascio E, Gerebtzoff G, Rodríguez-Pérez R. Systematic Evaluation of Local and Global Machine Learning Models for the Prediction of ADME Properties. Mol Pharm 2023; 20:1758-1767. [PMID: 36745394 DOI: 10.1021/acs.molpharmaceut.2c00962] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Machine learning (ML) has become an indispensable tool to predict absorption, distribution, metabolism, and excretion (ADME) properties in pharmaceutical research. ML algorithms are trained on molecular structures and corresponding ADME assay data to develop quantitative structure-property relationship (QSPR) models. Traditional QSPR models were trained on compound sets of limited size. With the advent of more complex ML algorithms and data availability, training sets have become larger and more diverse. Most common training approaches consist in either training a model with a small set of similar compounds, namely, compounds designed for the same drug discovery project or chemical series (local model approach) or with a larger set of diverse compounds (global model approach). Global models are built with all experimental data available for an assay, combining compound data from different projects and disease areas. Despite the ML progress made so far, the choice of the appropriate data composition for building ML models is still unclear. Herein, a systematic evaluation of local and global ML models was performed for 10 different experimental assays and 112 drug discovery projects. Results show a consistent superior performance of global models for ADME property predictions. Diagnostic analyses were also carried out to investigate the influence of training set size, structural diversity, and data shift in the relative performance of local and global ML models. Training set and structural diversity did not have an impact in the relative performance on the methods. Instead, data shift helped to identify the projects with larger performance differences between local and global models. Results presented in this work can be leveraged to improve ML-based ADME properties predictions and thus decision-making in drug discovery projects.
Collapse
Affiliation(s)
- Elena Di Lascio
- Novartis Institutes for Biomedical Research, Novartis Campus, BaselCH-4002, Switzerland
| | - Grégori Gerebtzoff
- Novartis Institutes for Biomedical Research, Novartis Campus, BaselCH-4002, Switzerland
| | | |
Collapse
|
14
|
Srikanth G, Ravi A, Sebastian A, Joseph J, Khanfar MA, El‐Gamal MI, Al‐Qawasmeh RA, Shehadi IA, McN. Sieburth S, Abu‐Yousef IA, Majdalawieh AF, Al‐Tel TH. Diastereoselective Synthesis of Camptothecin‐like Scaffolds: Construction of a New Class of Pseudo‐natural Products. European J Org Chem 2023. [DOI: 10.1002/ejoc.202300080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Affiliation(s)
- Gourishetty Srikanth
- Department of Biology Chemistry and Environmental Sciences American University of Sharjah 26666 Sharjah United Arab Emirates
| | - Anil Ravi
- Sharjah Institute for Medical research University of Sharjah P.O. Box 27272 Sharjah United Arab Emirates
| | - Anusha Sebastian
- Sharjah Institute for Medical research University of Sharjah P.O. Box 27272 Sharjah United Arab Emirates
| | - Jobi Joseph
- Sharjah Institute for Medical research University of Sharjah P.O. Box 27272 Sharjah United Arab Emirates
| | - Monther A. Khanfar
- College of Science Department of Chemistry University of Sharjah 27272 Sharjah United Arab Emirates
| | - Mohammed I. El‐Gamal
- Sharjah Institute for Medical research University of Sharjah P.O. Box 27272 Sharjah United Arab Emirates
- College of Pharmacy University of Sharjah P.O. Box 27272 Sharjah United Arab Emirates
| | - Raed A. Al‐Qawasmeh
- College of Science Department of Chemistry University of Sharjah 27272 Sharjah United Arab Emirates
| | - Ihsan A. Shehadi
- College of Science Department of Chemistry University of Sharjah 27272 Sharjah United Arab Emirates
| | - Scott McN. Sieburth
- Department of Chemistry Temple University 201 Beury Hall 19122 Philadelphia PA USA
| | - Imad A. Abu‐Yousef
- Department of Biology Chemistry and Environmental Sciences American University of Sharjah 26666 Sharjah United Arab Emirates
| | - Amin F. Majdalawieh
- Department of Biology Chemistry and Environmental Sciences American University of Sharjah 26666 Sharjah United Arab Emirates
| | - Taleb H. Al‐Tel
- Sharjah Institute for Medical research University of Sharjah P.O. Box 27272 Sharjah United Arab Emirates
- College of Pharmacy University of Sharjah P.O. Box 27272 Sharjah United Arab Emirates
| |
Collapse
|
15
|
Vivek-Ananth R, Sahoo AK, Baskaran SP, Samal A. Scaffold and Structural Diversity of the Secondary Metabolite Space of Medicinal Fungi. ACS OMEGA 2023; 8:3102-3113. [PMID: 36713723 PMCID: PMC9878629 DOI: 10.1021/acsomega.2c06428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/10/2022] [Indexed: 06/18/2023]
Abstract
Medicinal fungi, including mushrooms, have well-documented therapeutic uses. In this study, we perform a cheminformatics-based investigation of the scaffold and structural diversity of the secondary metabolite space of medicinal fungi and, moreover, perform a detailed comparison with approved drugs, other natural product libraries, and semi-synthetic libraries. We find that the secondary metabolite space of medicinal fungi has similar or higher scaffold diversity in comparison to other natural product libraries analyzed here. Notably, 94% of the scaffolds in the secondary metabolite space of medicinal fungi are not present in the approved drugs. Further, we find that the secondary metabolites, on the one hand, are structurally far from the approved drugs, while, on the other hand, they are close in terms of molecular properties to the approved drugs. Lastly, chemical space visualization using dimensionality reduction methods showed that the secondary metabolite space has minimal overlap with the approved drug space. In a nutshell, our results underscore that the secondary metabolite space of medicinal fungi is a valuable resource for identifying potential lead molecules for natural product-based drug discovery.
Collapse
Affiliation(s)
- R.P. Vivek-Ananth
- The
Institute of Mathematical Sciences (IMSc), Chennai600113, India
- Homi
Bhabha National Institute (HBNI), Mumbai400094, India
| | - Ajaya Kumar Sahoo
- The
Institute of Mathematical Sciences (IMSc), Chennai600113, India
- Homi
Bhabha National Institute (HBNI), Mumbai400094, India
| | - Shanmuga Priya Baskaran
- The
Institute of Mathematical Sciences (IMSc), Chennai600113, India
- Homi
Bhabha National Institute (HBNI), Mumbai400094, India
| | - Areejit Samal
- The
Institute of Mathematical Sciences (IMSc), Chennai600113, India
- Homi
Bhabha National Institute (HBNI), Mumbai400094, India
| |
Collapse
|
16
|
Comparative analysis of an anthraquinone and chalcone derivatives-based virtual combinatorial library. A cheminformatics "proof-of-concept" study. J Mol Graph Model 2022; 117:108307. [PMID: 36096064 DOI: 10.1016/j.jmgm.2022.108307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 01/14/2023]
Abstract
A Laplacian scoring algorithm for gene selection and the Gini coefficient to identify the genes whose expression varied least across a large set of samples were the state-of-the-art methods used here. These methods have not been trialed for their feasibility in cheminformatics. This was a maiden attempt to investigate a complete comparative analysis of an anthraquinone and chalcone derivatives-based virtual combinatorial library. This computational "proof-of-concept" study illustrated the combinatorial approach used to explain how the structure of the selected natural products (NPs) undergoes molecular diversity analysis. A virtual combinatorial library (1.6 M) based on 20 anthraquinones and 24 chalcones was enumerated. The resulting compounds were optimized to the near drug-likeness properties, and the physicochemical descriptors were calculated for all datasets including FDA, Non-FDA, and NPs from ZINC 15. UMAP and PCA were applied to compare and represent the chemical space coverage of each dataset. Subsequently, the Laplacian score and Gini coefficient were applied to delineate feature selection and selectivity among properties, respectively. Finally, we demonstrated the diversity between the datasets by employing Murcko's and the central scaffolds systems, calculating three fingerprint descriptors and analyzing their diversity by PCA and SOM. The optimized enumeration resulted in 1,610,268 compounds with NP-Likeness, and synthetic feasibility mean scores close to FDA, Non-FDA, and NPs datasets. The overlap between the chemical space of the 1.6 M database was more prominent than with the NPs dataset. A Laplacian score prioritized NP-likeness and hydrogen bond acceptor properties (1.0 and 0.923), respectively, while the Gini coefficient showed that all properties have selective effects on datasets (0.81-0.93). Scaffold and fingerprint diversity indicated that the descending order for the tested datasets was FDA, Non-FDA, NPs and 1.6 M. Virtual combinatorial libraries based on NPs can be considered as a source of the combinatorial compound with NP-likeness properties. Furthermore, measuring molecular diversity is supposed to be performed by different methods to allow for comparison and better judgment.
Collapse
|
17
|
In Silico Drug Repurposing Framework Predicts Repaglinide, Agomelatine and Protokylol as TRPV1 Modulators with Analgesic Activity. Pharmaceutics 2022; 14:pharmaceutics14122563. [PMID: 36559057 PMCID: PMC9781017 DOI: 10.3390/pharmaceutics14122563] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
Pain is one of the most common symptoms experienced by patients. The use of current analgesics is limited by low efficacy and important side effects. Transient receptor potential vanilloid-1 (TRPV1) is a non-selective cation channel, activated by capsaicin, heat, low pH or pro-inflammatory agents. Since TRPV1 is a potential target for the development of novel analgesics due to its distribution and function, we aimed to develop an in silico drug repositioning framework to predict potential TRPV1 ligands among approved drugs as candidates for treating various types of pain. Structures of known TRPV1 agonists and antagonists were retrieved from ChEMBL databases and three datasets were established: agonists, antagonists and inactive molecules (pIC50 or pEC50 < 5 M). Structures of candidates for repurposing were retrieved from the DrugBank database. The curated active/inactive datasets were used to build and validate ligand-based predictive models using Bemis−Murcko structural scaffolds, plain ring systems, flexophore similarities and molecular descriptors. Further, molecular docking studies were performed on both active and inactive conformations of the TRPV1 channel to predict the binding affinities of repurposing candidates. Variables obtained from calculated scaffold-based activity scores, molecular descriptors criteria and molecular docking were used to build a multi-class neural network as an integrated machine learning algorithm to predict TRPV1 antagonists and agonists. The proposed predictive model had a higher accuracy for classifying TRPV1 agonists than antagonists, the ROC AUC values being 0.980 for predicting agonists, 0.972 for antagonists and 0.952 for inactive molecules. After screening the approved drugs with the validated algorithm, repaglinide (antidiabetic) and agomelatine (antidepressant) emerged as potential TRPV1 antagonists, and protokylol (bronchodilator) as an agonist. Further studies are required to confirm the predicted activity on TRPV1 and to assess the candidates’ efficacy in alleviating pain.
Collapse
|
18
|
Zabolotna Y, Bonachera F, Horvath D, Lin A, Marcou G, Klimchuk O, Varnek A. Chemspace Atlas: Multiscale Chemography of Ultralarge Libraries for Drug Discovery. J Chem Inf Model 2022; 62:4537-4548. [DOI: 10.1021/acs.jcim.2c00509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Yuliana Zabolotna
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081, France
| | - Fanny Bonachera
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081, France
| | - Dragos Horvath
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081, France
| | - Arkadii Lin
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081, France
| | - Gilles Marcou
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081, France
| | - Olga Klimchuk
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081, France
| | - Alexandre Varnek
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081, France
| |
Collapse
|
19
|
Novel 5-Nitrofuran-Tagged Imidazo-Fused Azines and Azoles Amenable by the Groebke–Blackburn–Bienaymé Multicomponent Reaction: Activity Profile against ESKAPE Pathogens and Mycobacteria. Biomedicines 2022; 10:biomedicines10092203. [PMID: 36140307 PMCID: PMC9496245 DOI: 10.3390/biomedicines10092203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/26/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
A chemically diverse set of 13 5-nitrofuran-tagged heterocyclic compounds has been prepared via the Groebke–Blackburn–Bienaymé multicomponent reaction. The testing of these compounds against the so-called ESKAPE panel of pathogens identified an apparent lead compound—N-cyclohexyl-2-(5-nitrofuran-2-yl)imidazo[1,2-a]pyridine-3-amine (4a)—which showed an excellent profile against Enterobacter cloacae, Staphylococcus aureus, Klebsiella pneumoniae, and Enterococcus faecalis (MIC 0.25, 0.06, 0.25 and 0.25 µg/mL, respectively). Its antibacterial profile and practically convenient synthesis warrant further pre-clinical development. Certain structure-activity relationships were established in the course of this study which were rationalized by the flexible docking experiments in silico. The assessment of antitubercular potential of the compounds synthesized against drug sensitive H37v strain of Mycobacterium tuberculosis revealed little potential of the imidazo-fused products of the Groebke–Blackburn–Bienaymé multicomponent reaction as chemotherapeutic agents against this pathogen.
Collapse
|
20
|
Moshawih S, Goh HP, Kifli N, Idris AC, Yassin H, Kotra V, Goh KW, Liew KB, Ming LC. Synergy between machine learning and natural products cheminformatics: Application to the lead discovery of anthraquinone derivatives. Chem Biol Drug Des 2022; 100:185-217. [PMID: 35490393 DOI: 10.1111/cbdd.14062] [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: 01/18/2022] [Revised: 04/15/2022] [Accepted: 04/23/2022] [Indexed: 11/28/2022]
Abstract
Cheminformatics utilizing machine learning (ML) techniques have opened up a new horizon in drug discovery. This is owing to vast chemical space expansion with rocketing numbers of expected hits and lead compounds that match druggable macromolecular targets, in particular from natural compounds. Due to the natural products' (NP) structural complexity, uniqueness, and diversity, they could occupy a bigger space in pharmaceuticals, allowing the industry to pursue more selective leads in the nanomolar range of binding affinity. ML is an essential part of each step of the drug design pipeline, such as target prediction, compound library preparation, and lead optimization. Notably, molecular mechanic and dynamic simulations, induced docking, and free energy perturbations are essential in predicting best binding poses, binding free energy values, and molecular mechanics force fields. Those applications have leveraged from artificial intelligence (AI), which decreases the computational costs required for such costly simulations. This review aimed to describe chemical space and compound libraries related to NPs. High-throughput screening utilized for fractionating NPs and high-throughput virtual screening and their strategies, and significance, are reviewed. Particular emphasis was given to AI approaches, ML tools, algorithms, and techniques, especially in drug discovery of macrocyclic compounds and approaches in computer-aided and ML-based drug discovery. Anthraquinone derivatives were discussed as a source of new lead compounds that can be developed using ML tools for diverse medicinal uses such as cancer, infectious diseases, and metabolic disorders. Furthermore, the power of principal component analysis in understanding relevant protein conformations, and molecular modeling of protein-ligand interaction were also presented. Apart from being a concise reference for cheminformatics, this review is a useful text to understand the application of ML-based algorithms to molecular dynamics simulation and in silico absorption, distribution, metabolism, excretion, and toxicity prediction.
Collapse
Affiliation(s)
- Said Moshawih
- PAP Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Hui Poh Goh
- PAP Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Nurolaini Kifli
- PAP Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Azam Che Idris
- Faculty of Integrated Technologies, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Hayati Yassin
- Faculty of Integrated Technologies, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Vijay Kotra
- Faculty of Pharmacy, Quest International University, Perak, Malaysia
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai, Malaysia
| | - Kai Bin Liew
- Faculty of Pharmacy, University of Cyberjaya, Cyberjaya, Malaysia
| | - Long Chiau Ming
- PAP Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| |
Collapse
|
21
|
Zahra Sadeghian ZS, Bayat M. Synthesis of Heterocyclic Compounds Based on Isatins. CURR ORG CHEM 2022. [DOI: 10.2174/1385272826666220430145522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Isatin (1H-indole-2,3-diones) and its derivatives are a unique structure of heterocyclic molecules with great synthetic versatility and enormous biological activities of interest. Isatins have been broadly used as building blocks for the formation of a wide range of N-heterocycles. These applicable compounds undergo various reactions to form new heterocyclic compounds. The focus of this review is to summarize the recent literature and key reactions published about Pfitzinger, ring-opening, and ring expansion reactions of isatin and its derivatives during the period from 2018 to 2020. We believe this gives some insight and helps to bring about new ideas for further research.
Collapse
Affiliation(s)
| | - Mohammad Bayat
- Department of Chemistry, Faculty of Science, Imam Khomeini International University, Qazvin, Iran
| |
Collapse
|
22
|
Townley C, McMurray L, Marsden SP, Nelson A. A unified "top-down" approach for the synthesis of diverse lead-like molecular scaffolds. Bioorg Med Chem Lett 2022; 62:128631. [PMID: 35181466 DOI: 10.1016/j.bmcl.2022.128631] [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: 01/04/2022] [Revised: 02/14/2022] [Accepted: 02/14/2022] [Indexed: 11/16/2022]
Abstract
A "top-down" synthetic approach enabled the step-efficient synthesis of 21 diverse novel molecular scaffolds. The scaffolds were derived from four complex intermediates that had been prepared using cycloaddition chemistry. Scaffold-hopping of these intermediates was achieved through attachment of an additional ring, ring cleavage, ring expansion and/or ring fusion. It was shown that the resulting scaffolds could be decorated to yield diverse lead-like screening compounds.
Collapse
Affiliation(s)
- Chloe Townley
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK; Oncology R&D, AstraZeneca, Cambridge, CB4 0WG, United Kingdom
| | - Lindsay McMurray
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK; Oncology R&D, AstraZeneca, Cambridge, CB4 0WG, United Kingdom
| | - Stephen P Marsden
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK; Oncology R&D, AstraZeneca, Cambridge, CB4 0WG, United Kingdom
| | - Adam Nelson
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK; Oncology R&D, AstraZeneca, Cambridge, CB4 0WG, United Kingdom
| |
Collapse
|
23
|
Guntner AS, Bögl T, Mlynek F, Buchberger W. Large-Scale Evaluation of Collision Cross Sections to Investigate Blood-Brain Barrier Permeation of Drugs. Pharmaceutics 2021; 13:pharmaceutics13122141. [PMID: 34959422 PMCID: PMC8703848 DOI: 10.3390/pharmaceutics13122141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/30/2021] [Accepted: 12/08/2021] [Indexed: 11/16/2022] Open
Abstract
Successful drug administration to the central nervous system requires accurate adjustment of the drugs’ molecular properties. Therefore, structure-derived descriptors of potential brain therapeutic agents are essential for an early evaluation of pharmacokinetics during drug development. The collision cross section (CCS) of molecules was recently introduced as a novel measurable parameter to describe blood-brain barrier (BBB) permeation. This descriptor combines molecular information about mass, structure, volume, branching and flexibility. As these chemical properties are known to influence cerebral pharmacokinetics, CCS determination of new drug candidates may provide important additional spatial information to support existing models of BBB penetration of drugs. Besides measuring CCS, calculation is also possible; but however, the reliability of computed CCS values for an evaluation of BBB permeation has not yet been fully investigated. In this work, prediction tools based on machine learning were used to compute CCS values of a large number of compounds listed in drug libraries as negative or positive with respect to brain penetration (BBB+ and BBB− compounds). Statistical evaluation of computed CCS and several other descriptors could prove the high value of CCS. Further, CCS-deduced maximum molecular size of BBB+ drugs matched the dimensions of BBB pores. A threshold for transcellular penetration and possible permeation through pore-like openings of cellular tight-junctions is suggested. In sum, CCS evaluation with modern in silico tools shows high potential for its use in the drug development process.
Collapse
Affiliation(s)
- Armin Sebastian Guntner
- Institute of Analytical and General Chemistry, Johannes Kepler University, 4040 Linz, Austria
| | - Thomas Bögl
- Institute of Analytical and General Chemistry, Johannes Kepler University, 4040 Linz, Austria
| | - Franz Mlynek
- Institute of Analytical and General Chemistry, Johannes Kepler University, 4040 Linz, Austria
| | - Wolfgang Buchberger
- Institute of Analytical and General Chemistry, Johannes Kepler University, 4040 Linz, Austria
| |
Collapse
|
24
|
Zabolotna Y, Volochnyuk DM, Ryabukhin SV, Gavrylenko K, Horvath D, Klimchuk O, Oksiuta O, Marcou G, Varnek A. SynthI: A New Open-Source Tool for Synthon-Based Library Design. J Chem Inf Model 2021; 62:2151-2163. [PMID: 34723532 DOI: 10.1021/acs.jcim.1c00754] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Most of the existing computational tools for de novo library design are focused on the generation, rational selection, and combination of promising structural motifs to form members of the new library. However, the absence of a direct link between the chemical space of the retrosynthetically generated fragments and the pool of available reagents makes such approaches appear as rather theoretical and reality-disconnected. In this context, here we present Synthons Interpreter (SynthI), a new open-source toolkit for de novo library design that allows merging those two chemical spaces into a single synthons space. Here synthons are defined as actual fragments with valid valences and special labels, specifying the position and the nature of reactive centers. They can be issued from either the "breakup" of reference compounds according to 38 retrosynthetic rules or real reagents, after leaving group withdrawal or transformation. Such an approach not only enables the design of synthetically accessible libraries and analog generation but also facilitates reagents (building blocks) analysis in the medicinal chemistry context. SynthI code is publicly available at https://github.com/Laboratoire-de-Chemoinformatique/SynthI.
Collapse
Affiliation(s)
- Yuliana Zabolotna
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081, France
| | - Dmitriy M Volochnyuk
- Institute of Organic Chemistry, National Academy of Sciences of Ukraine, Murmanska Street 5, Kyiv 02660, Ukraine.,Enamine Ltd.78 Chervonotkatska str., 02660 Kyiv, Ukraine
| | - Sergey V Ryabukhin
- The Institute of High Technologies, Kyiv National Taras Shevchenko University, 64 Volodymyrska Street, Kyiv 01601, Ukraine.,Enamine Ltd.78 Chervonotkatska str., 02660 Kyiv, Ukraine
| | - Kostiantyn Gavrylenko
- Research-And-Education ChemBioCenter, National Taras Shevchenko University of Kyiv, Chervonotkatska str., 61, 03022 Kyiv, Ukraine.,Enamine Ltd.78 Chervonotkatska str., 02660 Kyiv, Ukraine
| | - Dragos Horvath
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081, France
| | - Olga Klimchuk
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081, France
| | - Oleksandr Oksiuta
- Institute of Organic Chemistry, National Academy of Sciences of Ukraine, Murmanska Street 5, Kyiv 02660, Ukraine.,Chemspace, Chervonotkatska Street 78, 02094 Kyiv, Ukraine
| | - Gilles Marcou
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081, France
| | - Alexandre Varnek
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081, France.,Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Kita 21 Nishi 10, Kita-ku, 001-0021 Sapporo, Japan
| |
Collapse
|
25
|
Lever J, Brkljača R, Rix C, Urban S. Application of Networking Approaches to Assess the Chemical Diversity, Biogeography, and Pharmaceutical Potential of Verongiida Natural Products. Mar Drugs 2021; 19:582. [PMID: 34677481 PMCID: PMC8539549 DOI: 10.3390/md19100582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/06/2023] Open
Abstract
This study provides a review of all isolated natural products (NPs) reported for sponges within the order Verongiida (1960 to May 2020) and includes a comprehensive compilation of their geographic and physico-chemical parameters. Physico-chemical parameters were used in this study to infer pharmacokinetic properties as well as the potential pharmaceutical potential of NPs from this order of marine sponge. In addition, a network analysis for the NPs produced by the Verongiida sponges was applied to systematically explore the chemical space relationships between taxonomy, secondary metabolite and drug score variables, allowing for the identification of differences and correlations within a dataset. The use of scaffold networks as well as bipartite relationship networks provided a platform to explore chemical diversity as well as the use of chemical similarity networks to link pharmacokinetic properties with structural similarity. This study paves the way for future applications of network analysis procedures in the field of natural products for any order or family.
Collapse
Affiliation(s)
- James Lever
- School of Science (Applied Chemistry and Environmental Sciences), RMIT University, GPO Box 2476, Melbourne, VIC 3001, Australia; (J.L.); (C.R.)
| | - Robert Brkljača
- Monash Biomedical Imaging, Monash University, Clayton, VIC 3168, Australia;
| | - Colin Rix
- School of Science (Applied Chemistry and Environmental Sciences), RMIT University, GPO Box 2476, Melbourne, VIC 3001, Australia; (J.L.); (C.R.)
| | - Sylvia Urban
- School of Science (Applied Chemistry and Environmental Sciences), RMIT University, GPO Box 2476, Melbourne, VIC 3001, Australia; (J.L.); (C.R.)
| |
Collapse
|
26
|
Ertl P. Magic Rings: Navigation in the Ring Chemical Space Guided by the Bioactive Rings. J Chem Inf Model 2021; 62:2164-2170. [PMID: 34445865 DOI: 10.1021/acs.jcim.1c00761] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The large majority of bioactive molecules contain a more or less complex ring system as a central structural element. This central core determines the basic molecule shape, keeps substituents in their proper positions, and often also contributes to the biological activity itself. In this study the ring systems extracted from one billion molecules are processed and differences between rings from bioactive molecules and common synthetic molecules are analyzed. The bioactive rings seem to be distributed throughout the large portion of chemical space, but not uniformly; one can see several more dense regions, where the bioactive rings often appear in small clusters, as well as empty areas. A web tool offering an interactive navigation in the ring chemical space and supporting identification of bioisosteric ring analogs available at https://bit.ly/magicrings is also described.
Collapse
Affiliation(s)
- Peter Ertl
- Novartis Institutes for BioMedical Research, CH-4056 Basel, Switzerland
| |
Collapse
|
27
|
Exploring the Effect of Structure-Based Scaffold Hopping on the Inhibition of Coxsackievirus A24v Transduction by Pentavalent N-Acetylneuraminic Acid Conjugates. Int J Mol Sci 2021; 22:ijms22168418. [PMID: 34445134 PMCID: PMC8395083 DOI: 10.3390/ijms22168418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/26/2021] [Accepted: 07/31/2021] [Indexed: 11/17/2022] Open
Abstract
Coxsackievirus A24 variant (CVA24v) is the primary causative agent of the highly contagious eye infection designated acute hemorrhagic conjunctivitis (AHC). It is solely responsible for two pandemics and several recurring outbreaks of the disease over the last decades, thus affecting millions of individuals throughout the world. To date, no antiviral agents or vaccines are available for combating this disease, and treatment is mainly supportive. CVA24v utilizes Neu5Ac-containing glycans as attachment receptors facilitating entry into host cells. We have previously reported that pentavalent Neu5Ac conjugates based on a glucose-scaffold inhibit CVA24v infection of human corneal epithelial cells. In this study, we report on the design and synthesis of scaffold-replaced pentavalent Neu5Ac conjugates and their effect on CVA24v cell transduction and the use of cryogenic electron microscopy (cryo-EM) to study the binding of these multivalent conjugates to CVA24v. The results presented here provide insights into the development of Neu5Ac-based inhibitors of CVA24v and, most significantly, the first application of cryo-EM to study the binding of a multivalent ligand to a lectin.
Collapse
|
28
|
Karageorgis G, Foley DJ, Laraia L, Brakmann S, Waldmann H. Pseudo Natural Products-Chemical Evolution of Natural Product Structure. Angew Chem Int Ed Engl 2021; 60:15705-15723. [PMID: 33644925 PMCID: PMC8360037 DOI: 10.1002/anie.202016575] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/27/2021] [Indexed: 01/05/2023]
Abstract
Pseudo-natural products (PNPs) combine natural product (NP) fragments in novel arrangements not accessible by current biosynthesis pathways. As such they can be regarded as non-biogenic fusions of NP-derived fragments. They inherit key biological characteristics of the guiding natural product, such as chemical and physiological properties, yet define small molecule chemotypes with unprecedented or unexpected bioactivity. We iterate the design principles underpinning PNP scaffolds and highlight their syntheses and biological investigations. We provide a cheminformatic analysis of PNP collections assessing their molecular properties and shape diversity. We propose and discuss how the iterative analysis of NP structure, design, synthesis, and biological evaluation of PNPs can be regarded as a human-driven branch of the evolution of natural products, that is, a chemical evolution of natural product structure.
Collapse
Affiliation(s)
- George Karageorgis
- Max-Planck Institute of Molecular PhysiologyOtto-Hahn Strasse 1144227DortmundGermany
| | - Daniel J. Foley
- Max-Planck Institute of Molecular PhysiologyOtto-Hahn Strasse 1144227DortmundGermany
- Current address: School of Physical and Chemical SciencesUniversity of CanterburyPrivate Bag 4800Christchurch8140New Zealand
| | - Luca Laraia
- Max-Planck Institute of Molecular PhysiologyOtto-Hahn Strasse 1144227DortmundGermany
- Current address: Department of ChemistryTechnical University of Denmark, kemitorvet 2072800 Kgs.LyngbyDenmark
| | - Susanne Brakmann
- Faculty of Chemistry and Chemical BiologyTU Dortmund UniversityOtto-Hahn Strasse 4a44227DortmundGermany
| | - Herbert Waldmann
- Max-Planck Institute of Molecular PhysiologyOtto-Hahn Strasse 1144227DortmundGermany
- Faculty of Chemistry and Chemical BiologyTU Dortmund UniversityOtto-Hahn Strasse 4a44227DortmundGermany
| |
Collapse
|
29
|
Iusupov IR, Curreli F, Spiridonov EA, Markov PO, Ahmed S, Belov DS, Manasova EV, Altieri A, Kurkin AV, Debnath AK. Design of gp120 HIV-1 entry inhibitors by scaffold hopping via isosteric replacements. Eur J Med Chem 2021; 224:113681. [PMID: 34246921 DOI: 10.1016/j.ejmech.2021.113681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
We present the development of alternative scaffolds and validation of their synthetic pathways as a tool for the exploration of new HIV gp120 inhibitors based on the recently discovered inhibitor of this class, NBD-14136. The new synthetic routes were based on isosteric replacements of the amine and acid precursors required for the synthesis of NBD-14136, guided by molecular modeling and chemical feasibility analysis. To ensure that these synthetic tools and new scaffolds had the potential for further exploration, we eventually tested few representative compounds from each newly designed scaffold against the gp120 inhibition assay and cell viability assays.
Collapse
Affiliation(s)
- Ildar R Iusupov
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Francesca Curreli
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, 10065, New York, United States
| | - Evgeniy A Spiridonov
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Pavel O Markov
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Shahad Ahmed
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, 10065, New York, United States
| | - Dmitry S Belov
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Ekaterina V Manasova
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Andrea Altieri
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia.
| | - Alexander V Kurkin
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia.
| | - Asim K Debnath
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, 10065, New York, United States.
| |
Collapse
|
30
|
Umedera K, Morita T, Yoshimori A, Yamada K, Katoh A, Kouji H, Nakamura H. Synthesis of Three-Dimensional (Di)Azatricyclododecene Scaffold and Its Application to Peptidomimetics. Chemistry 2021; 27:11888-11894. [PMID: 34060167 DOI: 10.1002/chem.202101440] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Indexed: 11/07/2022]
Abstract
A novel sp3 carbon-rich tricyclic 3D scaffold-based peptide mimetic compound library was constructed to target protein-protein interactions. Tricyclic framework 7 was synthesized from 9-azabicyclo[3,3,1]nonan-3-one (11) via a gold(I)-catalyzed Conia-ene reaction. The electron-donating group on the pendant alkyne of cyclization precursor 12 b-e was the key to forming 6-endo-dig cyclized product 7 with complete regioselectivity. Using the synthetic strategy for regioselective construction of bridged tricyclic framework 7, a diazatricyclododecene 3D-scaffold 8 a, which enables the introduction of substituents into the scaffold to mimic amino acid side chains, was designed and synthesized. The peptide mimetics 21 a-u were synthesized via step-by-step installation of three substituents on diazatricyclododecene scaffold 8 a. Compounds 21 a-h were synthesized as α-helix peptide mimics of hydrophobic ZZxxZ and ZxxZZ sequences (Z=Leu or Phe) and subjected to cell-based assays: antiproliferative activity, HIF-1 transcriptional activity which is considered to affect cancer malignancy, and antiviral activity against rabies virus. Compound 21 a showed the strongest inhibitory activity of HIF-1 transcriptional activity (IC50 =4.1±0.8 μM), whereas compounds 21 a-g showed antiviral activity with IC50 values of 4.2-12.4 μM, suggesting that the 3D-scaffold 8 a has potential as a versatile peptide mimic skeleton.
Collapse
Affiliation(s)
- Kohei Umedera
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8503, Japan
| | - Taiki Morita
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8503, Japan.,Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, 226-8503, Japan
| | - Atsushi Yoshimori
- Institute for Theoretical Medicine, Inc., 26-1, Muraoka-Higashi 2-chome, Fujisawa, 251-0012, Japan
| | - Kentaro Yamada
- Faculty of Agriculture Department of Veterinary Sciences, University of Miyazaki, Miyazaki, 889-2192, Japan.,Faculty of Medicine, Oita University, 1-1, Idaigaoka, Hasama-machi, Yufu-city, Oita, 879-5593, Japan
| | - Akira Katoh
- Faculty of Medicine, Oita University, 1-1, Idaigaoka, Hasama-machi, Yufu-city, Oita, 879-5593, Japan.,Institute of Advanced Medcine, Inc., Oita University, 17-20, Higashi kasuga-machi, Oita-city, Oita, 870-0037, Japan
| | - Hiroyuki Kouji
- Faculty of Medicine, Oita University, 1-1, Idaigaoka, Hasama-machi, Yufu-city, Oita, 879-5593, Japan.,Institute of Advanced Medcine, Inc., Oita University, 17-20, Higashi kasuga-machi, Oita-city, Oita, 870-0037, Japan
| | - Hiroyuki Nakamura
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8503, Japan.,Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, 226-8503, Japan
| |
Collapse
|
31
|
Chhabra S, Kumar S, Parkesh R. Chemical Space Exploration of DprE1 Inhibitors Using Chemoinformatics and Artificial Intelligence. ACS OMEGA 2021; 6:14430-14441. [PMID: 34124465 PMCID: PMC8190903 DOI: 10.1021/acsomega.1c01314] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/23/2021] [Indexed: 05/27/2023]
Abstract
Tuberculosis (TB), entrained by Mycobacterium tuberculosis, continues to be an enfeebling disease, killing nearly 1.5 million people in 2019, with 2 billion people worldwide affected by latent TB. The multidrug-resistant and totally drug-resistant emerging strains further exacerbate the TB infection. The cell wall of bacteria provides critical virulence components such as cell surface proteins, regulators, signal transduction proteins, and toxins. The cell wall biosynthesis pathway of Mycobacterium tuberculosis is exhaustively studied to discover novel drug targets. Decaprenylphosphoryl-β-d-ribose-2'-epimerase (DprE1) is an important enzyme involved in the arabinogalactan biosynthetic pathway of Mycobacterium tuberculosis cell wall and is essential for both latent and persistent bacterial infection. We analyzed all known ∼1300 DprE1 inhibitors to gain deep insights into the chemogenomic space of DprE1-ligand complexes. Physicochemical descriptors of the DprE1 inhibitors showed a marked lipophilic character forming a cluster distinct from the existing TB drugs, as revealed by the principal component analysis. Similarity analysis using Murcko scaffolds and rubber band scaling revealed scarce representation of the chemical space. Further, Murcko scaffold analysis uncovered favorable and unfavorable scaffolds, where benzo and pyridine-based core scaffolds exhibit the highest biological activity, as evidenced by their MIC and IC50 values. Automatic SAR and R-group decomposition analysis resulted in the identification of substructures responsible for the inhibitory activity of the DprE1 enzyme. Further, with activity cliff analysis, we observed prominent discontinuity in the SAR of DprE1 inhibitors, where even simple structural modification in the chemical scaffold resulted in significant potency difference, presumably due to the binding orientation and interaction in the active site. Thiophene, 6-membered aromatic rings, and unsubstituted benzene ring-based toxicophores were identified in the DprE1 chemical space using an artificial intelligence approach based on inductive logic programming. This paper, hence, ushers in new insights for the design and development of potent covalent and non-covalent DprE1 inhibitors and guides hit and lead optimization for the development of non-hazardous small molecule therapeutics for Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Sonali Chhabra
- CSIR-Institute
of Microbial Technology, Chandigarh 160036, India
- Academy
of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Sunil Kumar
- CSIR-Institute
of Microbial Technology, Chandigarh 160036, India
| | - Raman Parkesh
- CSIR-Institute
of Microbial Technology, Chandigarh 160036, India
- Academy
of Scientific and Innovative Research, Ghaziabad 201002, India
| |
Collapse
|
32
|
Karageorgis G, Foley DJ, Laraia L, Brakmann S, Waldmann H. Pseudo Natural Products—Chemical Evolution of Natural Product Structure. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202016575] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- George Karageorgis
- Max-Planck Institute of Molecular Physiology Otto-Hahn Strasse 11 44227 Dortmund Germany
| | - Daniel J. Foley
- Max-Planck Institute of Molecular Physiology Otto-Hahn Strasse 11 44227 Dortmund Germany
- Current address: School of Physical and Chemical Sciences University of Canterbury Private Bag 4800 Christchurch 8140 New Zealand
| | - Luca Laraia
- Max-Planck Institute of Molecular Physiology Otto-Hahn Strasse 11 44227 Dortmund Germany
- Current address: Department of Chemistry Technical University of Denmark, kemitorvet 207 2800 Kgs. Lyngby Denmark
| | - Susanne Brakmann
- Faculty of Chemistry and Chemical Biology TU Dortmund University Otto-Hahn Strasse 4a 44227 Dortmund Germany
| | - Herbert Waldmann
- Max-Planck Institute of Molecular Physiology Otto-Hahn Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical Biology TU Dortmund University Otto-Hahn Strasse 4a 44227 Dortmund Germany
| |
Collapse
|
33
|
Nelson A, Karageorgis G. Natural product-informed exploration of chemical space to enable bioactive molecular discovery. RSC Med Chem 2021; 12:353-362. [PMID: 34046620 PMCID: PMC8130614 DOI: 10.1039/d0md00376j] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/08/2020] [Indexed: 12/27/2022] Open
Abstract
The search for new bioactive molecules remains an open challenge limiting our ability to discover new drugs to treat disease and chemical probes to comprehensively study biological processes. The vastness of chemical space renders its exploration unfeasible by synthesis alone. Historically, chemists have tended to explore chemical space unevenly without committing to systematic frameworks for navigation. This minireview covers a range of approaches that take inspiration from the structure or origin of natural products, and help focus molecular discovery on biologically-relevant regions of chemical space. All these approaches have enabled the discovery of distinctive and novel bioactive small molecules such as useful chemical probes of biological mechanisms. This minireview comments on how such approaches may be developed into more general frameworks for the systematic identification of currently unexplored regions of biologically-relevant chemical space, a challenge that is central to both chemical biology and medicinal chemistry.
Collapse
Affiliation(s)
- Adam Nelson
- School of Chemistry, University of Leeds Woodhouse Lane LS2 9JT UK
- Astbury Centre for Structural and Molecular Biology, University of Leeds Woodhouse Lane LS2 9JT UK
| | - George Karageorgis
- School of Chemistry, University of Leeds Woodhouse Lane LS2 9JT UK
- Astbury Centre for Structural and Molecular Biology, University of Leeds Woodhouse Lane LS2 9JT UK
| |
Collapse
|
34
|
Makara GM, Kovács L, Szabó I, Pőcze G. Derivatization Design of Synthetically Accessible Space for Optimization: In Silico Synthesis vs Deep Generative Design. ACS Med Chem Lett 2021; 12:185-194. [PMID: 33603964 DOI: 10.1021/acsmedchemlett.0c00540] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/18/2020] [Indexed: 12/25/2022] Open
Abstract
Molecular design is of utmost importance in lead optimization programs ultimately determining the fate of the project and the speed to reach preclinical stage. Newly designed lead analogues or new chemotypes must successfully address the challenges in the multidimensional optimization process throughout several optimization cycles. The speed, quality, and creativity of the designs can have a major impact on the cycle time, the number of required cycles, and the number of compounds needed to be synthesized and evaluated that in combination affect the overall timeline and cost of the lead optimization phase. Recently, a new concept, generative design with deep learning, has become popular for de novo design of project relevant analogue sets. We have developed a de novo design technology called "derivatization design" that applies artificial-intelligence-assisted forward in silico synthesis for the generation of near neighbor lead analogues as well as scaffold variations. The several attractive features of the methodology include synthetic feasibility, reagent availability and cost data associated with each new molecule; thus, detailed synthetic assessment is automatically generated during the design. As a result, these practically important data types can become an early part of the ranking and selection process for cycle time reduction. The power of derivatization design is demonstrated in a simple design study of DDR1 inhibitors and comparison of the produced molecules to a recently published data set obtained with deep generative design.
Collapse
Affiliation(s)
| | | | - István Szabó
- ChemPass Ltd., 7 Záhony St, Budapest 1031, Hungary
| | - Gábor Pőcze
- ChemPass Ltd., 7 Záhony St, Budapest 1031, Hungary
| |
Collapse
|
35
|
Rice S, Cox DJ, Marsden SP, Nelson A. Efficient unified synthesis of diverse bridged polycyclic scaffolds using a complexity-generating 'stitching' annulation approach. Chem Commun (Camb) 2021; 57:599-602. [PMID: 33345263 DOI: 10.1039/d0cc06975b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Regioselective and stereospecific directed C-H arylation of simple amine substrates, and cyclisation, delivered 30 diverse, three-dimensional scaffolds. The unified approach significantly expanded the range of bridged ring systems that contain both a nitrogen atom and an aromatic ring.
Collapse
Affiliation(s)
- Scott Rice
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK. and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Daniel J Cox
- Redbrick Molecular, The Innovation Centre, 217 Portobello, Sheffield, S1 4DP, UK
| | | | - Adam Nelson
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK. and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
36
|
Kumar A, Loharch S, Kumar S, Ringe RP, Parkesh R. Exploiting cheminformatic and machine learning to navigate the available chemical space of potential small molecule inhibitors of SARS-CoV-2. Comput Struct Biotechnol J 2020; 19:424-438. [PMID: 33391634 PMCID: PMC7771909 DOI: 10.1016/j.csbj.2020.12.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 12/16/2022] Open
Abstract
The current life-threatening and tenacious pandemic eruption of coronavirus disease in 2019 (COVID-19) has posed a significant global hazard concerning high mortality rate, economic meltdown, and everyday life distress. The rapid spread of COVID-19 demands countermeasures to combat this deadly virus. Currently, there are no drugs approved by the FDA to treat COVID-19. Therefore, discovering small molecule therapeutics for treating COVID-19 infection is essential. So far, only a few small molecule inhibitors are reported for coronaviruses. There is a need to expand the small chemical space of coronaviruses inhibitors by adding potent and selective scaffolds with anti-COVID activity. In this context, the huge antiviral chemical space already available can be analysed using cheminformatic and machine learning to unearth new scaffolds. We created three specific datasets called "antiviral dataset" (N = 38,428) "drug-like antiviral dataset" (N = 20,963) and "anticorona dataset" (N = 433) for this purpose. We analyzed the 433 molecules of "anticorona dataset" for their scaffold diversity, physicochemical distributions, principal component analysis, activity cliffs, R-group decomposition, and scaffold mapping. The scaffold diversity of the "anticorona dataset" in terms of Murcko scaffold analysis demonstrates a thorough representation of diverse chemical scaffolds. However, physicochemical descriptor analysis and principal component analysis demonstrated negligible drug-like features for the "anticorona dataset" molecules. The "antiviral dataset" and "drug-like antiviral dataset" showed low scaffold diversity as measured by the Gini coefficient. The hierarchical clustering of the "antiviral dataset" against the "anticorona dataset" demonstrated little molecular similarity. We generated a library of frequent fragments and polypharmacological ligands targeting various essential viral proteins such as main protease, helicase, papain-like protease, and replicase polyprotein 1ab. Further structural and chemical features of the "anticorona dataset" were compared with SARS-CoV-2 repurposed drugs, FDA-approved drugs, natural products, and drugs currently in clinical trials. Using machine learning tool DCA (DMax Chemistry Assistant), we converted the "anticorona dataset" into an elegant hypothesis with significant functional biological relevance. Machine learning analysis uncovered that FDA approved drugs, Tizanidine HCl, Cefazolin, Raltegravir, Azilsartan, Acalabrutinib, Luliconazole, Sitagliptin, Meloxicam (Mobic), Succinyl sulfathiazole, Fluconazole, and Pranlukast could be repurposed as effective drugs for COVID-19. Fragment-based scaffold analysis and R-group decomposition uncovered pyrrolidine and the indole molecular scaffolds as the potent fragments for designing and synthesizing the novel drug-like molecules for targeting SARS-CoV-2. This comprehensive and systematic assessment of small-molecule viral therapeutics' entire chemical space realised critical insights to potentially privileged scaffolds that could aid in enrichment and rapid discovery of efficacious antiviral drugs for COVID-19.
Collapse
Affiliation(s)
- Abhinit Kumar
- GNRPC, CSIR – Institute of Microbial Technology, Chandigarh - 160036, India
| | - Saurabh Loharch
- GNRPC, CSIR – Institute of Microbial Technology, Chandigarh - 160036, India
| | - Sunil Kumar
- GNRPC, CSIR – Institute of Microbial Technology, Chandigarh - 160036, India
| | - Rajesh P. Ringe
- GNRPC, CSIR – Institute of Microbial Technology, Chandigarh - 160036, India
| | - Raman Parkesh
- GNRPC, CSIR – Institute of Microbial Technology, Chandigarh - 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad - 201002, India
| |
Collapse
|
37
|
Coley CW, Eyke NS, Jensen KF. Autonome Entdeckung in den chemischen Wissenschaften, Teil II: Ausblick. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201909989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Connor W. Coley
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Natalie S. Eyke
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Klavs F. Jensen
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| |
Collapse
|
38
|
Mervin LH, Johansson S, Semenova E, Giblin KA, Engkvist O. Uncertainty quantification in drug design. Drug Discov Today 2020; 26:474-489. [PMID: 33253918 DOI: 10.1016/j.drudis.2020.11.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/13/2020] [Accepted: 11/23/2020] [Indexed: 01/03/2023]
Abstract
Machine learning and artificial intelligence are increasingly being applied to the drug-design process as a result of the development of novel algorithms, growing access, the falling cost of computation and the development of novel technologies for generating chemically and biologically relevant data. There has been recent progress in fields such as molecular de novo generation, synthetic route prediction and, to some extent, property predictions. Despite this, most research in these fields has focused on improving the accuracy of the technologies, rather than on quantifying the uncertainty in the predictions. Uncertainty quantification will become a key component in autonomous decision making and will be crucial for integrating machine learning and chemistry automation to create an autonomous design-make-test-analyse cycle. This review covers the empirical, frequentist and Bayesian approaches to uncertainty quantification, and outlines how they can be used for drug design. We also outline the impact of uncertainty quantification on decision making.
Collapse
Affiliation(s)
- Lewis H Mervin
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK.
| | - Simon Johansson
- Department of Computer Science and Engineering, Chalmers University of Technology, Gothenburg, Sweden; Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Elizaveta Semenova
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Kathryn A Giblin
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Ola Engkvist
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
39
|
Medina-Franco JL, Saldívar-González FI. Cheminformatics to Characterize Pharmacologically Active Natural Products. Biomolecules 2020; 10:E1566. [PMID: 33213003 PMCID: PMC7698493 DOI: 10.3390/biom10111566] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/11/2020] [Accepted: 11/14/2020] [Indexed: 12/19/2022] Open
Abstract
Natural products have a significant role in drug discovery. Natural products have distinctive chemical structures that have contributed to identifying and developing drugs for different therapeutic areas. Moreover, natural products are significant sources of inspiration or starting points to develop new therapeutic agents. Natural products such as peptides and macrocycles, and other compounds with unique features represent attractive sources to address complex diseases. Computational approaches that use chemoinformatics and molecular modeling methods contribute to speed up natural product-based drug discovery. Several research groups have recently used computational methodologies to organize data, interpret results, generate and test hypotheses, filter large chemical databases before the experimental screening, and design experiments. This review discusses a broad range of chemoinformatics applications to support natural product-based drug discovery. We emphasize profiling natural product data sets in terms of diversity; complexity; acid/base; absorption, distribution, metabolism, excretion, and toxicity (ADME/Tox) properties; and fragment analysis. Novel techniques for the visual representation of the chemical space are also discussed.
Collapse
Affiliation(s)
- José L. Medina-Franco
- DIFACQUIM Research Group, Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Mexico City 04510, Mexico;
| | | |
Collapse
|
40
|
Mervin LH, Afzal AM, Engkvist O, Bender A. Comparison of Scaling Methods to Obtain Calibrated Probabilities of Activity for Protein–Ligand Predictions. J Chem Inf Model 2020; 60:4546-4559. [DOI: 10.1021/acs.jcim.0c00476] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Lewis H. Mervin
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Avid M. Afzal
- Data Sciences & Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Ola Engkvist
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Mölndal SE-431 83, Sweden
| | - Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge CB2 1TN, U.K
| |
Collapse
|
41
|
Coley CW, Eyke NS, Jensen KF. Autonomous Discovery in the Chemical Sciences Part II: Outlook. Angew Chem Int Ed Engl 2020; 59:23414-23436. [PMID: 31553509 DOI: 10.1002/anie.201909989] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Indexed: 01/19/2023]
Abstract
This two-part Review examines how automation has contributed to different aspects of discovery in the chemical sciences. In this second part, we reflect on a selection of exemplary studies. It is increasingly important to articulate what the role of automation and computation has been in the scientific process and how that has or has not accelerated discovery. One can argue that even the best automated systems have yet to "discover" despite being incredibly useful as laboratory assistants. We must carefully consider how they have been and can be applied to future problems of chemical discovery in order to effectively design and interact with future autonomous platforms. The majority of this Review defines a large set of open research directions, including improving our ability to work with complex data, build empirical models, automate both physical and computational experiments for validation, select experiments, and evaluate whether we are making progress towards the ultimate goal of autonomous discovery. Addressing these practical and methodological challenges will greatly advance the extent to which autonomous systems can make meaningful discoveries.
Collapse
Affiliation(s)
- Connor W Coley
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Natalie S Eyke
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Klavs F Jensen
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
42
|
Yang Y, Zhang Y, Hua Y, Chen X, Fan Y, Wang Y, Liang L, Deng C, Lu T, Chen Y, Liu H. In Silico Design and Analysis of a Kinase-Focused Combinatorial Library Considering Diversity and Quality. J Chem Inf Model 2020; 60:92-107. [PMID: 31886658 DOI: 10.1021/acs.jcim.9b00841] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A structurally diverse, high-quality, and kinase-focused database plays a critical role in finding hits or leads in kinase drug discovery. Here, we propose a workflow for designing a virtual kinase-focused combinatorial library using existing structures. Based on the analysis of known protein kinase inhibitors (PKIs), detailed fragment optimization, fragment selection, fragment linking, and a molecular filtering scheme were defined. Quick recognition of core fragments that can possibly form dual hydrogen bonds with the hinge region of the ATP-pocket was proposed. Furthermore, three diversity and four quality metrics were chosen for compound library analysis, which can be applied to databases with over 30 million structures. Compared with 13 commercial libraries, our protocol demonstrates a special advantage in terms of good skeleton diversity, acceptable fingerprint diversity, balanced scaffold distribution, and high quality, which can work well not only on existing PKIs, but also on four chosen commercial libraries. Overall, the strategy can greatly facilitate the expansion of a desirable chemical space for kinase drug discovery.
Collapse
Affiliation(s)
- Yan Yang
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Yanmin Zhang
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Yi Hua
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Xingye Chen
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Yuanrong Fan
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Yuchen Wang
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Li Liang
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Chenglong Deng
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Tao Lu
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China.,State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| | - Haichun Liu
- Laboratory of Molecular Design and Drug Discovery, School of Science , China Pharmaceutical University , 639 Longmian Avenue , Nanjing 211198 , China
| |
Collapse
|
43
|
Lipkus AH, Watkins SP, Gengras K, McBride MJ, Wills TJ. Recent Changes in the Scaffold Diversity of Organic Chemistry As Seen in the CAS Registry. J Org Chem 2019; 84:13948-13956. [DOI: 10.1021/acs.joc.9b02111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Alan H. Lipkus
- CAS, P.O. Box 3012, Columbus, Ohio 43210-0012, United States
| | | | - Keith Gengras
- CAS, P.O. Box 3012, Columbus, Ohio 43210-0012, United States
| | | | - Todd J. Wills
- CAS, P.O. Box 3012, Columbus, Ohio 43210-0012, United States
| |
Collapse
|
44
|
Naveja JJ, Pilón-Jiménez BA, Bajorath J, Medina-Franco JL. A general approach for retrosynthetic molecular core analysis. J Cheminform 2019; 11:61. [PMID: 33430974 PMCID: PMC6760108 DOI: 10.1186/s13321-019-0380-5] [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: 03/04/2019] [Accepted: 08/04/2019] [Indexed: 11/13/2022] Open
Abstract
Scaffold analysis of compound data sets has reemerged as a chemically interpretable alternative to machine learning for chemical space and structure–activity relationships analysis. In this context, analog series-based scaffolds (ASBS) are synthetically relevant core structures that represent individual series of analogs. As an extension to ASBS, we herein introduce the development of a general conceptual framework that considers all putative cores of molecules in a compound data set, thus softening the often applied “single molecule–single scaffold” correspondence. A putative core is here defined as any substructure of a molecule complying with two basic rules: (a) the size of the core is a significant proportion of the whole molecule size and (b) the substructure can be reached from the original molecule through a succession of retrosynthesis rules. Thereafter, a bipartite network consisting of molecules and cores can be constructed for a database of chemical structures. Compounds linked to the same cores are considered analogs. We present case studies illustrating the potential of the general framework. The applications range from inter- and intra-core diversity analysis of compound data sets, structure–property relationships, and identification of analog series and ASBS. The molecule–core network herein presented is a general methodology with multiple applications in scaffold analysis. New statistical methods are envisioned that will be able to draw quantitative conclusions from these data. The code to use the method presented in this work is freely available as an additional file. Follow-up applications include analog searching and core structure–property relationships analyses.![]()
Collapse
Affiliation(s)
- J Jesús Naveja
- PECEM, School of Medicine, Universidad Nacional Autónoma de México, Avenida Universidad 3000, 04510, Mexico City, Mexico. .,Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Avenida Universidad 3000, 04510, Mexico City, Mexico.
| | - B Angélica Pilón-Jiménez
- Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Avenida Universidad 3000, 04510, Mexico City, Mexico
| | - Jürgen Bajorath
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, 53115, Bonn, Germany
| | - José L Medina-Franco
- Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Avenida Universidad 3000, 04510, Mexico City, Mexico.
| |
Collapse
|
45
|
Computational Drug Repurposing Algorithm Targeting TRPA1 Calcium Channel as a Potential Therapeutic Solution for Multiple Sclerosis. Pharmaceutics 2019; 11:pharmaceutics11090446. [PMID: 31480671 PMCID: PMC6781306 DOI: 10.3390/pharmaceutics11090446] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/19/2019] [Accepted: 08/24/2019] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease affecting the central nervous system (CNS) through neurodegeneration and demyelination, leading to physical/cognitive disability and neurological defects. A viable target for treating MS appears to be the Transient Receptor Potential Ankyrin 1 (TRPA1) calcium channel, whose inhibition has been shown to have beneficial effects on neuroglial cells and protect against demyelination. Using computational drug discovery and data mining methods, we performed an in silico screening study combining chemical graph mining, quantitative structure-activity relationship (QSAR) modeling, and molecular docking techniques in a global prediction model in order to identify repurposable drugs as potent TRPA1 antagonists that may serve as potential treatments for MS patients. After screening the DrugBank database with the combined generated algorithm, 903 repurposable structures were selected, with 97 displaying satisfactory inhibition probabilities and pharmacokinetics. Among the top 10 most probable inhibitors of TRPA1 with good blood brain barrier (BBB) permeability, desvenlafaxine, paliperidone, and febuxostat emerged as the most promising repurposable agents for treating MS. Molecular docking studies indicated that desvenlafaxine, paliperidone, and febuxostat are likely to induce allosteric TRPA1 channel inhibition. Future in vitro and in vivo studies are needed to confirm the biological activity of the selected hit molecules.
Collapse
|
46
|
Schoene J, Gazzi T, Lindemann P, Christmann M, Volkamer A, Nazaré M. Probing 2
H
‐Indazoles as Templates for SGK1, Tie2, and SRC Kinase Inhibitors. ChemMedChem 2019; 14:1514-1527. [DOI: 10.1002/cmdc.201900328] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/26/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Jens Schoene
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus BerlinBuch Robert-Roessle-Str. 10 13125 Berlin Germany
| | - Thais Gazzi
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus BerlinBuch Robert-Roessle-Str. 10 13125 Berlin Germany
| | - Peter Lindemann
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus BerlinBuch Robert-Roessle-Str. 10 13125 Berlin Germany
| | - Mathias Christmann
- Organische ChemieInstitut für Chemie und BiochemieFreie Universität Berlin Takustrasse. 3 14195 Berlin Germany
| | - Andrea Volkamer
- In silico Toxicology and Structural Bioinformatics Group, Institute of PhysiologyCharité—Universitätsmedizin Berlin Charitéplatz 1 10117 Berlin Germany
| | - Marc Nazaré
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus BerlinBuch Robert-Roessle-Str. 10 13125 Berlin Germany
- Anna-Louisa-Karsch-Str. 2 10178 Berlin Germany
| |
Collapse
|
47
|
Zhao C, Huang D, Li R, Xu Y, Su S, Gu Q, Xu J. Identifying Novel Anti-Osteoporosis Leads with a Chemotype-Assembly Approach. J Med Chem 2019; 62:5885-5900. [DOI: 10.1021/acs.jmedchem.9b00517] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Chao Zhao
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Dane Huang
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
- Guangdong Province Engineering Technology Research Institute of T.C.M., Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou 510095, China
| | - Ruyue Li
- Guangdong Province Engineering Technology Research Institute of T.C.M., Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou 510095, China
| | - Yida Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Shimin Su
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Qiong Gu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Jun Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
- School of Biotechnology and Health Sciences, Wuyi University, 99 Yingbin Road, Jiangmen 529020, China
| |
Collapse
|
48
|
Mayol-Llinàs J, Chow S, Nelson A. Expansion of the structure-activity relationships of BACE1 inhibitors by harnessing diverse building blocks prepared using a unified synthetic approach. MEDCHEMCOMM 2019; 10:616-620. [PMID: 31057741 PMCID: PMC6482882 DOI: 10.1039/c9md00085b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/14/2019] [Indexed: 11/21/2022]
Abstract
The structural diversity of β-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitors was expanded by harnessing diverse building blocks that had been prepared via a unified lead-oriented synthetic approach. It was shown that the lipophilic cyclohexylmethyl group within a known series of BACE1 inhibitors could be productively replaced with a range of alternative ring systems.
Collapse
Affiliation(s)
- Joan Mayol-Llinàs
- School of Chemistry , University of Leeds , Leeds , LS2 9JT , UK .
- Astbury Centre for Structural Molecular Biology , University of Leeds , Leeds , LS2 9JT , UK
| | - Shiao Chow
- School of Chemistry , University of Leeds , Leeds , LS2 9JT , UK .
- Astbury Centre for Structural Molecular Biology , University of Leeds , Leeds , LS2 9JT , UK
| | - Adam Nelson
- School of Chemistry , University of Leeds , Leeds , LS2 9JT , UK .
- Astbury Centre for Structural Molecular Biology , University of Leeds , Leeds , LS2 9JT , UK
| |
Collapse
|
49
|
Volochnyuk DM, Ryabukhin SV, Moroz YS, Savych O, Chuprina A, Horvath D, Zabolotna Y, Varnek A, Judd DB. Evolution of commercially available compounds for HTS. Drug Discov Today 2019; 24:390-402. [DOI: 10.1016/j.drudis.2018.10.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/02/2018] [Accepted: 10/30/2018] [Indexed: 12/17/2022]
|
50
|
Ashenden SK. Screening Library Design. Methods Enzymol 2018; 610:73-96. [PMID: 30390806 DOI: 10.1016/bs.mie.2018.09.016] [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: 02/18/2023]
Abstract
Thanks to technological advances and a greater understanding of the biological and chemical natures of targets and related diseases, high-throughput screening (HTS) has been allowed to be faster, cheaper, and more accessible. Yet, despite these increased technologies and understandings, the frequency of novel and drugs are being approved each year has not being increasing over the years. 2017 was considered a "bumper" year with a total of 46 approved drugs, over double that of the previous year. However, it is thought that part of the problem that HTS has not lived up to expectations is because of the contents of current chemical libraries. Therefore, new methods to design screening libraries are of great interest.
Collapse
Affiliation(s)
- Stephanie Kay Ashenden
- Department of Chemistry, Cambridge University, Cambridge, United Kingdom; Discovery Sciences, IMed Biotech Unit, AstraZeneca R&D, Cambridge, United Kingdom.
| |
Collapse
|