1
|
McGrath A, Huang H, Brazeau JF, Zhang Z, Audu CO, Vellore NA, Zhu L, Shi Z, Venable JD, Gelin CF, Cernak T. Modulating the Potency of BRD4 PROTACs at the Systems Level with Amine-Acid Coupling Reactions. J Med Chem 2024. [PMID: 39688565 DOI: 10.1021/acs.jmedchem.4c02047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Protein degradation using proteolysis targeting chimeras (PROTACs) represents a promising therapeutic strategy. PROTACs are heterobifunctional molecules that consist of a target-binding moiety and an E3 ligase binding moiety, connected by a linker. These fragments are frequently united via amide bonds. While straightforward to synthesize, amides may impart suboptimal drug properties to the overall molecule. From a systems level perspective, we envisioned that the potency of PROTACs could be modulated through selection of reaction conditions─wherein different catalysts produce distinct linkers from the same two building blocks. We present a suite of BRD4 PROTAC degraders prepared via four new amine-acid coupling reactions alongside the classic amide coupling. Our findings reveal that variations in reaction conditions affect the physicochemical properties of PROTACs, resulting in a spectrum of properties. Notably, several new PROTACs demonstrated enhanced BRD4 degradation efficacy compared to those employing amide linkers, emphasizing the potential of systems chemistry as a therapeutic optimization strategy.
Collapse
Affiliation(s)
- Andrew McGrath
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48104, United States
| | - Haiyan Huang
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48104, United States
| | - Jean-Francois Brazeau
- Therapeutics Discovery, Janssen Research & Development, LLC, La Jolla, California 92121, United States
| | - Zirong Zhang
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48104, United States
| | - Christopher O Audu
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48104, United States
| | - Nadeem A Vellore
- Therapeutics Discovery, Janssen Research & Development, LLC, La Jolla, California 92121, United States
| | - Lu Zhu
- Therapeutics Discovery, Janssen Research & Development, LLC, Spring House, Pennsylvania 19477, United States
| | - Zhicai Shi
- Therapeutics Discovery, Janssen Research & Development, LLC, Spring House, Pennsylvania 19477, United States
| | - Jennifer D Venable
- Therapeutics Discovery, Janssen Research & Development, LLC, La Jolla, California 92121, United States
| | - Christine F Gelin
- Therapeutics Discovery, Janssen Research & Development, LLC, La Jolla, California 92121, United States
| | - Tim Cernak
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48104, United States
| |
Collapse
|
2
|
Chen G, Yuan H, Zhang L, Zhang J, Li K, Wang X. Pancreatic lipase immobilization on cellulose filter paper for inhibitors screening and network pharmacology study of anti-obesity mechanism. Talanta 2024; 280:126750. [PMID: 39213890 DOI: 10.1016/j.talanta.2024.126750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/08/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The discovery of pancreatic lipase (PL) inhibitors is an essential route to develop new anti-obesity drugs. In this experiment, chitosan was used to add amino groups to cellulose filter paper (CFP) and then glutaraldehyde was used to covalently combine PL with amino-modified CFP through the Schiff base reaction. Under optimal immobilization conditions, CFP immobilized PL has a wide range of pH and temperature tolerance, as well as excellent reproducibility, reusability and storage stability. Subsequently, 26 natural products (NPs) were screened by immobilized PL with black tea extract having the highest inhibition rate. Three compounds with binding effects on PL (epigallocatechin gallate, theaflavin-3-gallate and theaflavin-3,3'-digallate) were captured. Molecular docking proved that these three compounds have a strong binding affinity for PL. Fluorescence spectra further revealed that theaflavin-3,3'-digallate could statically quench the intrinsic fluorescence of pancreatic lipase. The molecular docking and thermodynamic parameters indicated that electrostatic interaction was considered as the main interaction force between PL and theaflavin-3,3'-digallate. Finally, the potential anti-obesity targets and pathways of the three compounds were discussed through network pharmacology. This study not only proposes a simple and efficient method for screening PL inhibitors, but also sheds light on the anti-obesity mechanism of active compounds in black tea.
Collapse
Affiliation(s)
- Guangxuan Chen
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Huicong Yuan
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Lumei Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Jingran Zhang
- SCIEX, Analytical Instrument Trading Co., Ltd, Beijing, 100015, China
| | - Kefeng Li
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR, China.
| | - Xu Wang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, China.
| |
Collapse
|
3
|
Tredup C, Ackloo S, Beck H, Brown PJ, Bullock AN, Ciulli A, Dikic I, Edfeldt K, Edwards AM, Elkins JM, Farin HF, Fon EA, Gstaiger M, Günther J, Gustavsson AL, Häberle S, Isigkeit L, Huber KVM, Kotschy A, Krämer O, Leach AR, Marsden BD, Matsui H, Merk D, Montel F, Mulder MPC, Müller S, Owen DR, Proschak E, Röhm S, Stolz A, Sundström M, von Delft F, Willson TM, Arrowsmith CH, Knapp S. Toward target 2035: EUbOPEN - a public-private partnership to enable & unlock biology in the open. RSC Med Chem 2024:d4md00735b. [PMID: 39618964 PMCID: PMC11605244 DOI: 10.1039/d4md00735b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024] Open
Abstract
Target 2035 is a global initiative that seeks to identify a pharmacological modulator of most human proteins by the year 2035. As part of an ongoing series of annual updates of this initiative, we summarise here the efforts of the EUbOPEN project whose objectives and results are making a strong contribution to the goals of Target 2035. EUbOPEN is a public-private partnership with four pillars of activity: (1) chemogenomic library collections, (2) chemical probe discovery and technology development for hit-to-lead chemistry, (3) profiling of bioactive compounds in patient-derived disease assays, and (4) collection, storage and dissemination of project-wide data and reagents. The substantial outputs of this programme include a chemogenomic compound library covering one third of the druggable proteome, as well as 100 chemical probes, both profiled in patient derived assays, as well as hundreds of data sets deposited in existing public data repositories and a project-specific data resource for exploring EUbOPEN outputs.
Collapse
Affiliation(s)
- Claudia Tredup
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| | - Suzanne Ackloo
- Structural Genomics Consortium, University of Toronto - St George Campus 101 College Street, MaRS Center South Tower 7th Floor Toronto Canada
| | - Hartmut Beck
- Drug Discovery Sciences, Research & Development, Pharmaceuticals, Bayer AG Wuppertal Nordrhein-Westfalen Germany
| | - Peter J Brown
- Structural Genomics Consortium, University of North Carolina at Chapel Hill Campus Box 7356, 120 Mason Farm Road, GMB 1070 Chapel Hill North Carolina USA
| | - Alex N Bullock
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
| | - Alessio Ciulli
- Centre for Targeted Protein Degradation, University of Dundee, School of Life Sciences 1 James Lindsay Place DD1 5JJ Dundee UK
| | - Ivan Dikic
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty Frankfurt am Main Germany
- Buchmann Institute for Molecular Lifesciences, Goethe University Frankfurt Frankfurt am Main Germany
| | - Kristina Edfeldt
- Structural Genomics Consortium, Department of Medicine, Karolinska University Hospital and Karolinska Institutet Stockholm Sweden
| | - Aled M Edwards
- Structural Genomics Consortium, University of Toronto - St George Campus 101 College Street, MaRS Center South Tower 7th Floor Toronto Canada
| | - Jonathan M Elkins
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
| | - Henner F Farin
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy Frankfurt am Main Hessen Germany
| | - Edward A Fon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital (The Neuro), McGill University Montreal Canada
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology ETH Zürich Zurich ZH Switzerland
| | | | - Anna-Lena Gustavsson
- Chemical Biology Consortium Sweden, Department of Medical Biochemistry & Biophysics, Karolinska Institute Stockholm Sweden
| | - Sandra Häberle
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| | - Laura Isigkeit
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
| | - Kilian V M Huber
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
| | - Andras Kotschy
- Servier Research Institute of Medicinal Chemistry Budapest Hungary
| | - Oliver Krämer
- Discovery Research Coordination, Boehringer Ingelheim International GmbH Binger Straße 173 55216 Ingelheim am Rhein Germany
| | - Andrew R Leach
- European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus Hinxton Cambridge UK
| | - Brian D Marsden
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
| | - Hisanori Matsui
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited Fujisawa Kanagawa Japan
| | - Daniel Merk
- Ludwig-Maximilians-Universitat Munchen Munchen Germany
| | - Florian Montel
- Discovery Research Coordination, Boehringer Ingelheim Pharma GmbH & Co. KG Birkendorfer Straße 65 88397 Biberach an der Riss Germany
| | - Monique P C Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center Leiden The Netherlands
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| | | | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP Theodor-Stern-Kai 7 60596 Frankfurt am Main Germany
| | - Sandra Röhm
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| | - Alexandra Stolz
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty Frankfurt am Main Germany
- Buchmann Institute for Molecular Lifesciences, Goethe University Frankfurt Frankfurt am Main Germany
| | - Michael Sundström
- Structural Genomics Consortium, Department of Medicine, Karolinska University Hospital and Karolinska Institutet Stockholm Sweden
| | - Frank von Delft
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
- Diamond Light Source, Harwell Science and Innovation Campus Didcot OX11 0DE UK
| | - Timothy M Willson
- Structural Genomics Consortium, University of North Carolina at Chapel Hill Campus Box 7356, 120 Mason Farm Road, GMB 1070 Chapel Hill North Carolina USA
| | - Cheryl H Arrowsmith
- Structural Genomics Consortium, University of Toronto - St George Campus 101 College Street, MaRS Center South Tower 7th Floor Toronto Canada
- Princess Margaret Cancer Centre Toronto Ontario M5G 1L7 Canada
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| |
Collapse
|
4
|
Lescano LE, Salazar MO, Furlan RLE. Chemically engineered essential oils prepared through thiocyanation under solvent-free conditions: chemical and bioactivity alteration. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:35. [PMID: 38822174 PMCID: PMC11143095 DOI: 10.1007/s13659-024-00456-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/17/2024] [Indexed: 06/02/2024]
Abstract
The generation of chemically engineered essential oils (CEEOs) prepared from bi-heteroatomic reactions using ammonium thiocyanate as a source of bioactive compounds is described. The impact of the reaction on the chemical composition of the mixtures was qualitatively demonstrated through GC-MS, utilizing univariate and multivariate analysis. The reaction transformed most of the components in the natural mixtures, thereby expanding the chemical diversity of the mixtures. Changes in inhibition properties between natural and CEEOs were demonstrated through acetylcholinesterase TLC autography, resulting in a threefold increase in the number of positive events due to the modification process. The chemically engineered Origanum vulgare L. essential oil was subjected to bioguided fractionation, leading to the discovery of four new active compounds with similar or higher potency than eserine against the enzyme. The results suggest that the directed chemical transformation of essential oils can be a valuable strategy for discovering new acetylcholinesterase (AChE) inhibitors.
Collapse
Affiliation(s)
- Liz E Lescano
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Suipacha 531, 2000, Rosario, Argentina
| | - Mario O Salazar
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Suipacha 531, 2000, Rosario, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Suipacha 531, 2000, Rosario, Argentina.
| | - Ricardo L E Furlan
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Suipacha 531, 2000, Rosario, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Suipacha 531, 2000, Rosario, Argentina
| |
Collapse
|
5
|
Liang J, Wang Y, Liu B, Dong X, Cai W, Zhang N, Zhang H. Deciphering the intricate linkage between the gut microbiota and Alzheimer's disease: Elucidating mechanistic pathways promising therapeutic strategies. CNS Neurosci Ther 2024; 30:e14704. [PMID: 38584341 PMCID: PMC10999574 DOI: 10.1111/cns.14704] [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: 10/04/2023] [Revised: 12/15/2023] [Accepted: 03/25/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND The gut microbiome is composed of various microorganisms such as bacteria, fungi, and protozoa, and constitutes an important part of the human gut. Its composition is closely related to human health and disease. Alzheimer's disease (AD) is a neurodegenerative disease whose underlying mechanism has not been fully elucidated. Recent research has shown that there are significant differences in the gut microbiota between AD patients and healthy individuals. Changes in the composition of gut microbiota may lead to the development of harmful factors associated with AD. In addition, the gut microbiota may play a role in the development and progression of AD through the gut-brain axis. However, the exact nature of this relationship has not been fully understood. AIMS This review will elucidate the types and functions of gut microbiota and their relationship with AD and explore in depth the potential mechanisms of gut microbiota in the occurrence of AD and the prospects for treatment strategies. METHODS Reviewed literature from PubMed and Web of Science using key terminologies related to AD and the gut microbiome. RESULTS Research indicates that the gut microbiota can directly or indirectly influence the occurrence and progression of AD through metabolites, endotoxins, and the vagus nerve. DISCUSSION This review discusses the future challenges and research directions regarding the gut microbiota in AD. CONCLUSION While many unresolved issues remain regarding the gut microbiota and AD, the feasibility and immense potential of treating AD by modulating the gut microbiota are evident.
Collapse
Affiliation(s)
- Junyi Liang
- Heilongjiang University of Traditional Chinese MedicineHarbinHeilongjiang ProvinceChina
| | - Yueyang Wang
- Heilongjiang University of Traditional Chinese MedicineHarbinHeilongjiang ProvinceChina
| | - Bin Liu
- Heilongjiang University of Traditional Chinese MedicineHarbinHeilongjiang ProvinceChina
| | - Xiaohong Dong
- Jiamusi CollegeHeilongjiang University of Traditional Chinese MedicineJiamusiHeilongjiang ProvinceChina
| | - Wenhui Cai
- Heilongjiang University of Traditional Chinese MedicineHarbinHeilongjiang ProvinceChina
| | - Ning Zhang
- Heilongjiang University of Traditional Chinese MedicineHarbinHeilongjiang ProvinceChina
| | - Hong Zhang
- Heilongjiang Jiamusi Central HospitalJiamusiHeilongjiang ProvinceChina
| |
Collapse
|
6
|
Homer JA, Koelln RA, Barrow AS, Gialelis TL, Boiarska Z, Steinohrt NS, Lee EF, Yang WH, Johnson RM, Chung T, Habowski AN, Vishwakarma DS, Bhunia D, Avanzi C, Moorhouse AD, Jackson M, Tuveson DA, Lyons SK, Lukey MJ, Fairlie WD, Haider SM, Steinmetz MO, Prota AE, Moses JE. Modular synthesis of functional libraries by accelerated SuFEx click chemistry. Chem Sci 2024; 15:3879-3892. [PMID: 38487227 PMCID: PMC10935723 DOI: 10.1039/d3sc05729a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/09/2024] [Indexed: 03/17/2024] Open
Abstract
Accelerated SuFEx Click Chemistry (ASCC) is a powerful method for coupling aryl and alkyl alcohols with SuFEx-compatible functional groups. With its hallmark favorable kinetics and exceptional product yields, ASCC streamlines the synthetic workflow, simplifies the purification process, and is ideally suited for discovering functional molecules. We showcase the versatility and practicality of the ASCC reaction as a tool for the late-stage derivatization of bioactive molecules and in the array synthesis of sulfonate-linked, high-potency, microtubule targeting agents (MTAs) that exhibit nanomolar anticancer activity against multidrug-resistant cancer cell lines. These findings underscore ASCC's promise as a robust platform for drug discovery.
Collapse
Affiliation(s)
- Joshua A Homer
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Rebecca A Koelln
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Andrew S Barrow
- La Trobe Institute for Molecular Science, La Trobe University Melbourne VIC 3086 Australia
| | - Timothy L Gialelis
- La Trobe Institute for Molecular Science, La Trobe University Melbourne VIC 3086 Australia
| | - Zlata Boiarska
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut Villigen PSI 5232 Switzerland
- Department of Chemistry, Università degli Studi di Milano Via Golgi 19 20133 Milan Italy
| | - Nikita S Steinohrt
- Olivia Newton-John Cancer Research Institute Heidelberg Victoria 3084 Australia
- School of Cancer Medicine, La Trobe University Melbourne Victoria 3086 Australia
| | - Erinna F Lee
- Olivia Newton-John Cancer Research Institute Heidelberg Victoria 3084 Australia
- School of Cancer Medicine, La Trobe University Melbourne Victoria 3086 Australia
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University Melbourne Victoria 3086 Australia
| | - Wen-Hsuan Yang
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Robert M Johnson
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Taemoon Chung
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Amber N Habowski
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | | | - Debmalya Bhunia
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Charlotte Avanzi
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University Fort Collins CO 80523 USA
| | - Adam D Moorhouse
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University Fort Collins CO 80523 USA
| | - David A Tuveson
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Scott K Lyons
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Michael J Lukey
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - W Douglas Fairlie
- Olivia Newton-John Cancer Research Institute Heidelberg Victoria 3084 Australia
- School of Cancer Medicine, La Trobe University Melbourne Victoria 3086 Australia
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University Melbourne Victoria 3086 Australia
| | - Shozeb M Haider
- School of Pharmacy, University College London 29-39 Brunswick Square London WC1N 1AX UK
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut Villigen PSI 5232 Switzerland
- Biozentrum, University of Basel 4056 Basel Switzerland
| | - Andrea E Prota
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut Villigen PSI 5232 Switzerland
| | - John E Moses
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| |
Collapse
|
7
|
Karale UB, Shinde A, Gaikwad VR, Kalari S, Gourishetti K, Radhakrishnan M, Poornachandra Y, Amanchy R, Chakravarty S, Andugulapati SB, Rode HB. Iron mediated reductive cyclization/oxidation for the generation of chemically diverse scaffolds: An approach in drug discovery. Bioorg Chem 2023; 139:106698. [PMID: 37418784 DOI: 10.1016/j.bioorg.2023.106698] [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: 04/17/2023] [Revised: 06/03/2023] [Accepted: 06/20/2023] [Indexed: 07/09/2023]
Abstract
Chemically diverse scaffolds represent a main source of biologically important starting points in drug discovery. Herein, we report the development of such diverse scaffolds from nitroarene/ nitro(hetero)arenes using a key synthetic strategy. In a pilot-scale study, the synthesis of 10 diverse scaffolds was achieved. The 1,7-phenanthroline, thiazolo[5,4-f]quinoline, 2,3-dihydro-1H-pyrrolo[2,3-g]quinoline, pyrrolo[3,2-f]quinoline, 1H-[1,4]oxazino[3,2-g]quinolin-2(3H)-one, [1,2,5]oxadiazolo[3,4-h]quinoline, 7H-pyrido[2,3-c]carbazole, 3H-pyrazolo[4,3-f]quinoline, pyrido[3,2-f]quinoxaline were obtained from nitro hetero arenes in ethanol using iron-acetic acid treatment followed by reaction under oxygen atmosphere. This diverse library is compliant with the rule of five for drug-likeness. The mapping of chemical space represented by these scaffolds revealed a significant contribution to the underrepresented chemical diversity. Crucial to the development of this approach was the mapping of biological space covered by these scaffolds which revealed neurotropic and prophylactic anti-inflammatory activities. In vitro, neuro-biological assays revealed that compounds 14a and 15a showed excellent neurotropic potential and neurite growth compared to controls. Further, anti-inflammatory assays (in vitro and in vivo models) exhibited that Compound 16 showed significant anti-inflammatory activity by attenuating the LPS-induced TNF-α and CD68 levels by modulating the NFkB pathway. In addition, treatment with compound 16 significantly ameliorated the LPS-induced sepsis conditions, and pathological abnormalities (in lung and liver tissues) and improved the survival of the rats compared to LPS control. Owing to their chemical diversity along with bioactivities, it is envisaged that new quality pre-clinical candidates will be generated in the above therapeutic areas using identified leads.
Collapse
Affiliation(s)
- Uttam B Karale
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Akash Shinde
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India
| | - Vikas R Gaikwad
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India
| | - Saradhi Kalari
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Karthik Gourishetti
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India; Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500 007, India
| | - Mydhili Radhakrishnan
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India; Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500 007, India
| | - Yedla Poornachandra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500 007, India
| | - Ramars Amanchy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500 007, India
| | - Sumana Chakravarty
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India; Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500 007, India
| | - Sai Balaji Andugulapati
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India; Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500 007, India
| | - Haridas B Rode
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India.
| |
Collapse
|
8
|
Liang J, Liu B, Dong X, Wang Y, Cai W, Zhang N, Zhang H. Decoding the role of gut microbiota in Alzheimer's pathogenesis and envisioning future therapeutic avenues. Front Neurosci 2023; 17:1242254. [PMID: 37790586 PMCID: PMC10544353 DOI: 10.3389/fnins.2023.1242254] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/04/2023] [Indexed: 10/05/2023] Open
Abstract
Alzheimer's disease (AD) emerges as a perturbing neurodegenerative malady, with a profound comprehension of its underlying pathogenic mechanisms continuing to evade our intellectual grasp. Within the intricate tapestry of human health and affliction, the enteric microbial consortium, ensconced within the milieu of the human gastrointestinal tract, assumes a role of cardinal significance. Recent epochs have borne witness to investigations that posit marked divergences in the composition of the gut microbiota between individuals grappling with AD and those favored by robust health. The composite vicissitudes in the configuration of the enteric microbial assembly are posited to choreograph a participatory role in the inception and progression of AD, facilitated by the intricate conduit acknowledged as the gut-brain axis. Notwithstanding, the precise nature of this interlaced relationship remains enshrouded within the recesses of obscurity, poised for an exhaustive revelation. This review embarks upon the endeavor to focalize meticulously upon the mechanistic sway exerted by the enteric microbiota upon AD, plunging profoundly into the execution of interventions that govern the milieu of enteric microorganisms. In doing so, it bestows relevance upon the therapeutic stratagems that form the bedrock of AD's management, all whilst casting a prospective gaze into the horizon of medical advancements.
Collapse
Affiliation(s)
- Junyi Liang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Bin Liu
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Xiaohong Dong
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Yueyang Wang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Wenhui Cai
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Ning Zhang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Hong Zhang
- Heilongjiang Jiamusi Central Hospital, Jiamusi, Heilongjiang, China
| |
Collapse
|
9
|
Francis D, Farooque S, Meager A, Derks D, Leggott A, Warriner S, O'Neill AJ, Nelson A. Algorithm-driven activity-directed expansion of a series of antibacterial quinazolinones. Org Biomol Chem 2022; 20:9672-9678. [PMID: 36448404 DOI: 10.1039/d2ob01404a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Activity-directed synthesis (ADS) is a structure-blind, function driven approach that can drive the discovery of bioactive small molecules. In ADS, arrays of reactions are designed and executed, and the crude product mixtures are then directly screened to identify reactions that yield bioactive products. The design of subsequent reaction arrays is then informed by the hit reactions that are discovered. In this study, algorithms for reaction array design were developed in which the reactions to be executed were selected from a large set of virtual reactions; the reactions were selected on the basis of similarity to reactions known to yield bioactive products. The algorithms were harnessed to design arrays of photoredox-catalysed alkylation reactions whose crude products were then screened for inhibition of growth of S. aureus ATCC29213. It was demonstrated that the approach enabled expansion of a series of antibacterial quinazolinones. It is envisaged that such algorithms could ultimately enable fully autonomous activity-directed molecular discovery.
Collapse
Affiliation(s)
- Daniel Francis
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK. .,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Sannia Farooque
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK. .,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Archie Meager
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK. .,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Didi Derks
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK.
| | - Abbie Leggott
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK. .,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Stuart Warriner
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK. .,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Alex J O'Neill
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.,School of Molecular and Cellular 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
|
10
|
Li Z, Yao X, Zhang X, Mei H, Han J. Carboxylic Acid O-H Insertion Reaction of β-Ester Diazos Enabling Synthesis of β-Acyloxy Esters. J Org Chem 2022; 87:15483-15491. [PMID: 36354090 DOI: 10.1021/acs.joc.2c02023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Generation of non-stabilized β-ester diazos and their applications in carboxylic acid O-H insertion reactions have been reported, which afford β-acyloxy esters in excellent yield. Varieties of aryl- and alkyl-substituted diazos are well tolerated in this insertion reaction under mild and convenient conditions. Moreover, structural modification of the natural product and molecular drug can also be achieved in this reaction. This protocol not only realizes the reaction involving unstable β-ester diazos but also provides an efficient strategy for the synthesis of β-acyloxy esters.
Collapse
Affiliation(s)
- Ziyi Li
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Xinyu Yao
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Xin Zhang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Haibo Mei
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Jianlin Han
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
11
|
Anti-Alzheimer's disease potential of traditional chinese medicinal herbs as inhibitors of BACE1 and AChE enzymes. Biomed Pharmacother 2022; 154:113576. [PMID: 36007279 DOI: 10.1016/j.biopha.2022.113576] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease that often occurs in the elderly population. At present, most drugs for AD on the market are single-target drugs, which have achieved certain success in the treatment of AD. However, the efficacy and safety of single-target drugs have not achieved the expected results because AD is a multifactorial disease. Multi-targeted drugs act on multiple factors of the disease network to improve efficacy and reduce adverse reactions. Therefore, the search for effective dual-target or even multi-target drugs has become a new research trend. Many of results found that the dual-target inhibitors of the beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) and acetylcholinesterase (AChE) found from traditional Chinese medicine have a good inhibitory effect on AD with fewer side effects. This article reviews sixty-six compounds extracted from Chinese medicinal herbs, which have inhibitory activity on BACE1 and AChE. This provides a theoretical basis for the further development of these compounds as dual-target inhibitors for the treatment of AD.
Collapse
|
12
|
Tan G, Das M, Keum H, Bellotti P, Daniliuc C, Glorius F. Photochemical single-step synthesis of β-amino acid derivatives from alkenes and (hetero)arenes. Nat Chem 2022; 14:1174-1184. [PMID: 35915332 DOI: 10.1038/s41557-022-01008-w] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/23/2022] [Indexed: 11/09/2022]
Abstract
β-Amino acids are frequently found as important components in numerous biologically active molecules, drugs and natural products. In particular, they are broadly utilized in the construction of bioactive peptides and peptidomimetics, thanks to their increased metabolic stability. Despite the number of methodologies established for the preparation of β-amino acid derivatives, the majority of these methods require metal-mediated multistep manipulations of prefunctionalized substrates. Here we disclose a metal-free, energy-transfer enabled highly regioselective intermolecular aminocarboxylation reaction for the single-step installation of both amine and ester functionalities into alkenes or (hetero)arenes. A bifunctional oxime oxalate ester was developed to simultaneously generate C-centred ester and N-centred iminyl radicals. This mild method features a remarkably broad substrate scope (up to 140 examples) and excellent tolerance of sensitive functional groups, and substrates that range from the simplest ethylene to complex (hetero)arenes can participate in the reaction, thus offering a general and practical access to β-amino acid derivatives.
Collapse
Affiliation(s)
- Guangying Tan
- Westfälische Wilhelms-Universität Münster, Organisch-Chemisches Institut, Münster, Germany
| | - Mowpriya Das
- Westfälische Wilhelms-Universität Münster, Organisch-Chemisches Institut, Münster, Germany
| | - Hyeyun Keum
- Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science, Daejeon, South Korea.,Department of Chemistry, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Peter Bellotti
- Westfälische Wilhelms-Universität Münster, Organisch-Chemisches Institut, Münster, Germany
| | - Constantin Daniliuc
- Westfälische Wilhelms-Universität Münster, Organisch-Chemisches Institut, Münster, Germany
| | - Frank Glorius
- Westfälische Wilhelms-Universität Münster, Organisch-Chemisches Institut, Münster, Germany.
| |
Collapse
|
13
|
Thomas RP, Heap RE, Zappacosta F, Grant EK, Pogány P, Besley S, Fallon DJ, Hann MM, House D, Tomkinson NCO, Bush JT. A direct-to-biology high-throughput chemistry approach to reactive fragment screening. Chem Sci 2021; 12:12098-12106. [PMID: 34667575 PMCID: PMC8457371 DOI: 10.1039/d1sc03551g] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/05/2021] [Indexed: 11/21/2022] Open
Abstract
Methods for rapid identification of chemical tools are essential for the validation of emerging targets and to provide medicinal chemistry starting points for the development of new medicines. Here, we report a screening platform that combines 'direct-to-biology' high-throughput chemistry (D2B-HTC) with photoreactive fragments. The platform enabled the rapid synthesis of >1000 PhotoAffinity Bits (HTC-PhABits) in 384-well plates in 24 h and their subsequent screening as crude reaction products with a protein target without purification. Screening the HTC-PhABit library with carbonic anhydrase I (CAI) afforded 7 hits (0.7% hit rate), which were found to covalently crosslink in the Zn2+ binding pocket. A powerful advantage of the D2B-HTC screening platform is the ability to rapidly perform iterative design-make-test cycles, accelerating the development and optimisation of chemical tools and medicinal chemistry starting points with little investment of resource.
Collapse
Affiliation(s)
- Ross P Thomas
- GlaxoSmithKline Gunnels Wood Road Stevenage Hertfordshire SG1 2NY UK
- Department of Pure and Applied Chemistry, University of Strathclyde 295 Cathedral Street Glasgow G1 1XL UK
| | - Rachel E Heap
- GlaxoSmithKline South Collegeville Road Collegeville PA 19426 USA
| | | | - Emma K Grant
- GlaxoSmithKline Gunnels Wood Road Stevenage Hertfordshire SG1 2NY UK
| | - Peter Pogány
- GlaxoSmithKline Gunnels Wood Road Stevenage Hertfordshire SG1 2NY UK
| | - Stephen Besley
- GlaxoSmithKline Gunnels Wood Road Stevenage Hertfordshire SG1 2NY UK
| | - David J Fallon
- GlaxoSmithKline Gunnels Wood Road Stevenage Hertfordshire SG1 2NY UK
| | - Michael M Hann
- GlaxoSmithKline Gunnels Wood Road Stevenage Hertfordshire SG1 2NY UK
| | - David House
- GlaxoSmithKline Gunnels Wood Road Stevenage Hertfordshire SG1 2NY UK
| | - Nicholas C O Tomkinson
- Department of Pure and Applied Chemistry, University of Strathclyde 295 Cathedral Street Glasgow G1 1XL UK
| | - Jacob T Bush
- GlaxoSmithKline Gunnels Wood Road Stevenage Hertfordshire SG1 2NY UK
| |
Collapse
|
14
|
Zhou X, Tan Y, Gou K, Tao L, Luo Y, Zhou Y, Zuo Z, Sun Q, Luo Y, Zhao Y. Discovery of novel inhibitors of human phosphoglycerate dehydrogenase by activity-directed combinatorial chemical synthesis strategy. Bioorg Chem 2021; 115:105159. [PMID: 34298241 DOI: 10.1016/j.bioorg.2021.105159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 02/08/2023]
Abstract
Serine, the source of the one-carbon units essential for de novo purine and deoxythymidine synthesis plays a crucial role in the growth of cancer cells. Phosphoglycerate dehydrogenase (PHGDH) which catalyzes the first, rate-limiting step in de novo serine biosynthesis has become a promising target for the cancer treatment. Here we identified H-G6 as a potential PHGDH inhibitor from the screening of an in-house small molecule library based on the enzymatic assay. We adopted activity-directed combinatorial chemical synthesis strategy to optimize this hit compound. Compound b36 was found to be the noncompetitive and the most promising one with IC50 values of 5.96 ± 0.61 μM against PHGDH. Compound b36 inhibited the proliferation of human breast cancer and ovarian cancer cells, reduced intracellular serine synthesis, damaged DNA synthesis, and induced cell cycle arrest. Collectively, our results suggest that b36 is a novel PHGDH inhibitor, which could be a promising modulator to reprogram the serine synthesis pathway and might be a potential anticancer lead worth further exploration.
Collapse
Affiliation(s)
- Xia Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yuping Tan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Kun Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lei Tao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yuan Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yue Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zeping Zuo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Qingxiang Sun
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Youfu Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Yinglan Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
15
|
Wang Y, Wang YJ, Liang XC, Shen MH, Xu HD, Xu D. An aryl thiol-vinyl azide coupling reaction and a thiol-vinyl azide coupling/cyclization cascade: efficient synthesis of β-ketosulfides and arene-fused 5-methylene-2-pyrrolidinone derivatives. Org Biomol Chem 2021; 19:5169-5176. [PMID: 34037057 DOI: 10.1039/d1ob00328c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The addition reaction of thiol to vinyl azide has been extensively studied. Variously substituted aryl thiols are all viable for this coupling process. The scope of the other partner is successfully expanded from α-aryl vinyl azide to α-alkyl vinyl azide. A thiol-vinyl azide coupling/cyclization cascade is realized with substituted aryl vinyl azides carrying a 2-methoxycarbonyl group. The value of β-ketosulfide products was demonstrated by its application in S-heterocycle synthesis.
Collapse
Affiliation(s)
- Yong Wang
- Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province 213164, China.
| | - Yu-Jiao Wang
- Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province 213164, China.
| | - Xian-Chen Liang
- Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province 213164, China.
| | - Mei-Hua Shen
- Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province 213164, China.
| | - Hua-Dong Xu
- Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province 213164, China.
| | - Defeng Xu
- Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province 213164, China.
| |
Collapse
|
16
|
Grisoni F, Huisman BJH, Button AL, Moret M, Atz K, Merk D, Schneider G. Combining generative artificial intelligence and on-chip synthesis for de novo drug design. SCIENCE ADVANCES 2021; 7:eabg3338. [PMID: 34117066 PMCID: PMC8195470 DOI: 10.1126/sciadv.abg3338] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 04/23/2021] [Indexed: 05/24/2023]
Abstract
Automating the molecular design-make-test-analyze cycle accelerates hit and lead finding for drug discovery. Using deep learning for molecular design and a microfluidics platform for on-chip chemical synthesis, liver X receptor (LXR) agonists were generated from scratch. The computational pipeline was tuned to explore the chemical space of known LXRα agonists and generate novel molecular candidates. To ensure compatibility with automated on-chip synthesis, the chemical space was confined to the virtual products obtainable from 17 one-step reactions. Twenty-five de novo designs were successfully synthesized in flow. In vitro screening of the crude reaction products revealed 17 (68%) hits, with up to 60-fold LXR activation. The batch resynthesis, purification, and retesting of 14 of these compounds confirmed that 12 of them were potent LXR agonists. These results support the suitability of the proposed design-make-test-analyze framework as a blueprint for automated drug design with artificial intelligence and miniaturized bench-top synthesis.
Collapse
Affiliation(s)
- Francesca Grisoni
- ETH Zurich, Department of Chemistry and Applied Biosciences, RETHINK, Zurich, Switzerland.
- Eindhoven University of Technology, Department of Biomedical Engineering, Eindhoven, Netherlands
| | - Berend J H Huisman
- ETH Zurich, Department of Chemistry and Applied Biosciences, RETHINK, Zurich, Switzerland
| | - Alexander L Button
- ETH Zurich, Department of Chemistry and Applied Biosciences, RETHINK, Zurich, Switzerland
- University of Lausanne, Department of Computational Biology, Lausanne, Switzerland
| | - Michael Moret
- ETH Zurich, Department of Chemistry and Applied Biosciences, RETHINK, Zurich, Switzerland
| | - Kenneth Atz
- ETH Zurich, Department of Chemistry and Applied Biosciences, RETHINK, Zurich, Switzerland
| | - Daniel Merk
- ETH Zurich, Department of Chemistry and Applied Biosciences, RETHINK, Zurich, Switzerland.
- Goethe University Frankfurt, Institute of Pharmaceutical Chemistry, Frankfurt, Germany
| | - Gisbert Schneider
- ETH Zurich, Department of Chemistry and Applied Biosciences, RETHINK, Zurich, Switzerland.
- ETH Singapore SEC Ltd, Singapore, Singapore
| |
Collapse
|
17
|
Sharonova T, Paramonova P, Kalinin S, Bunev A, Gasanov RЕ, Nocentini A, Sharoyko V, Tennikova TB, Dar'in D, Supuran CT, Krasavin M. Insertion of metal carbenes into the anilinic N-H bond of unprotected aminobenzenesulfonamides delivers low nanomolar inhibitors of human carbonic anhydrase IX and XII isoforms. Eur J Med Chem 2021; 218:113352. [PMID: 33774343 DOI: 10.1016/j.ejmech.2021.113352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 01/25/2023]
Abstract
Herein we report the synthesis of a set of thirty-four primary sulfonamides generated via formal N-H-insertion of metal carbenes into anilinic amino group of sulfanilamide and its meta-substituted analog. Obtained compounds were tested in vitro as inhibitors of four physiologically significant isoforms of the metalloenzyme human carbonic anhydrase (hCA, EC 4.2.1.1). Many of the synthesized sulfonamides displayed low nanomolar Ki values against therapeutically relevant hCA II, IX, and XII, whereas they did not potently inhibit hCA I. Provided the promising activity profiles of the substances towards tumor-associated hCA IX and XII isozymes, single-concentration MTT test was performed for the entire set. Disappointingly, most of the discovered hCA inhibitors did not significantly suppress the growth of cancer cells either in normoxia or CoCl2 induced hypoxic conditions. The only two compounds exerting profound antiproliferative effect turned out to be modest hCA inhibitors. Their out of the range activity in cells is likely attributive to the presence of Michael acceptor substructure which can potentially act either through the inhibition of Thioredoxin reductases (TrxRs, EC 1.8.1.9) or nonspecific covalent binding to cell proteins.
Collapse
Affiliation(s)
- Tatiana Sharonova
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Polina Paramonova
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Stanislav Kalinin
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Alexander Bunev
- Medicinal Chemistry Center, Togliatti State University, Togliatti, 445020, Russian Federation
| | - Rovshan Е Gasanov
- Medicinal Chemistry Center, Togliatti State University, Togliatti, 445020, Russian Federation
| | - Alessio Nocentini
- Neurofarba Department, Universita degli Studi di Firenze, Florence, 50019, Italy
| | - Vladimir Sharoyko
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Tatiana B Tennikova
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Dmitry Dar'in
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Claudiu T Supuran
- Neurofarba Department, Universita degli Studi di Firenze, Florence, 50019, Italy.
| | - Mikhail Krasavin
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation.
| |
Collapse
|
18
|
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: 5.0] [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
|
19
|
Leggott A, Clarke JE, Chow S, Warriner SL, O'Neill AJ, Nelson A. Activity-directed expansion of a series of antibacterial agents. Chem Commun (Camb) 2021; 56:8047-8050. [PMID: 32538401 DOI: 10.1039/d0cc02361b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The feasibility of using activity-directed synthesis to drive antibacterial discovery was investigated. An array of 220 Pd-catalysed microscale reactions was executed, and the crude product mixtures were evaluated for activity against Staphylococcus aureus. Scale-up of the hit reactions, purification and evaluation, enabled expansion of a class of antibacterial quinazolinones. The novel antibacterials had MICs from 0.016 μg mL-1 (i.e. 38 nM) to 2-4 μg mL-1 against S. aureus ATCC29213.
Collapse
Affiliation(s)
- Abbie Leggott
- School of Chemistry, University of Leeds, LS2 9JT, Leeds, UK. and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| | - Justin E Clarke
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK. and School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Shiao Chow
- School of Chemistry, University of Leeds, LS2 9JT, Leeds, UK. and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| | - Stuart L Warriner
- School of Chemistry, University of Leeds, LS2 9JT, Leeds, UK. and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| | - Alex J O'Neill
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK. and School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Adam Nelson
- School of Chemistry, University of Leeds, LS2 9JT, Leeds, UK. and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
20
|
Green AI, Tinworth CP, Warriner S, Nelson A, Fey N. Computational Mapping of Dirhodium(II) Catalysts. Chemistry 2021; 27:2402-2409. [PMID: 32964545 PMCID: PMC7898874 DOI: 10.1002/chem.202003801] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/09/2020] [Indexed: 12/31/2022]
Abstract
The chemistry of dirhodium(II) catalysts is highly diverse, and can enable the synthesis of many different molecular classes. A tool to aid in catalyst selection, independent of mechanism and reactivity, would therefore be highly desirable. Here, we describe the development of a database for dirhodium(II) catalysts that is based on the principal component analysis of DFT-calculated parameters capturing their steric and electronic properties. This database maps the relevant catalyst space, and may facilitate exploration of the reactivity landscape for any process catalysed by dirhodium(II) complexes. We have shown that one of the principal components of these catalysts correlates with the outcome (e.g. yield, selectivity) of a transformation used in a molecular discovery project. Furthermore, we envisage that this approach will assist the selection of more effective catalyst screening sets, and, hence, the data-led optimisation of a wide range of rhodium-catalysed transformations.
Collapse
Affiliation(s)
- Adam I. Green
- School of Chemistry and Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsLS29JTUK
| | | | - Stuart Warriner
- School of Chemistry and Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsLS29JTUK
| | - Adam Nelson
- School of Chemistry and Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsLS29JTUK
| | - Natalie Fey
- School of ChemistryUniversity of BristolCantock's CloseBristolBS81TSUK
| |
Collapse
|
21
|
Karageorgis G, Liver S, Nelson A. Activity-Directed Synthesis: A Flexible Approach for Lead Generation. ChemMedChem 2020; 15:1776-1782. [PMID: 32734671 PMCID: PMC7589241 DOI: 10.1002/cmdc.202000524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Indexed: 11/06/2022]
Abstract
Activity-directed synthesis (ADS) is a structure-blind, functional-driven molecular discovery approach. In this Concept, four case studies highlight the general applicability of ADS and showcase its flexibility to support different medicinal chemistry strategies. ADS deliberately harnesses reactions with multiple possible outcomes, and allows many chemotypes to be evaluated in parallel. Resources are focused on bioactive molecules, which emerge in tandem with associated synthetic routes. Some of the future challenges for ADS are highlighted, including the realisation of an autonomous molecular discovery platform. The prospects for ADS to become a mainstream lead generation approach are discussed.
Collapse
Affiliation(s)
- George Karageorgis
- School of Chemistry and Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsLS2 9JTUK
| | - Samuel Liver
- School of Chemistry and Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsLS2 9JTUK
- Rosalind Franklin InstituteHarwell CampusDidcotOX11 0FAUK
| | - Adam Nelson
- School of Chemistry and Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsLS2 9JTUK
- Rosalind Franklin InstituteHarwell CampusDidcotOX11 0FAUK
| |
Collapse
|
22
|
Green AI, Hobor F, Tinworth CP, Warriner S, Wilson AJ, Nelson A. Activity-Directed Synthesis of Inhibitors of the p53/hDM2 Protein-Protein Interaction. Chemistry 2020; 26:10682-10689. [PMID: 32458465 PMCID: PMC7496268 DOI: 10.1002/chem.202002153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Indexed: 01/10/2023]
Abstract
Protein-protein interactions (PPIs) provide a rich source of potential targets for drug discovery and biomedical science research. However, the identification of structural-diverse starting points for discovery of PPI inhibitors remains a significant challenge. Activity-directed synthesis (ADS), a function-driven discovery approach, was harnessed in the discovery of the p53/hDM2 PPI. Over two rounds of ADS, 346 microscale reactions were performed, with prioritisation on the basis of the activity of the resulting product mixtures. Four distinct and novel series of PPI inhibitors were discovered that, through biophysical characterisation, were shown to have promising ligand efficiencies. It was thus shown that ADS can facilitate ligand discovery for a target that does not have a defined small-molecule binding site, and can provide distinctive starting points for the discovery of PPI inhibitors.
Collapse
Affiliation(s)
- Adam I. Green
- School of ChemistryUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
| | - Fruzsina Hobor
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- School of Molecular and Cellular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
| | | | - Stuart Warriner
- School of ChemistryUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
| | - Andrew J. Wilson
- School of ChemistryUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
| | - Adam Nelson
- School of ChemistryUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
| |
Collapse
|
23
|
Ueda J, Harada S, Kanda A, Nakayama H, Nemoto T. Silver-Catalyzed, Chemo- and Enantioselective Intramolecular Dearomatization of Indoles to Access Sterically Congested Azaspiro Frameworks. J Org Chem 2020; 85:10934-10950. [PMID: 32692554 DOI: 10.1021/acs.joc.0c01580] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
An asymmetric dearomatization of indoles bearing α-diazoacetamide functionalities was developed for synthesizing high-value spiro scaffolds. A silver phosphate chemoselectively catalyzed the sterically challenging dearomatization, whereas more typically used metal catalysts for carbene transfer reactions, such as a rhodium complex, were not effective and instead resulted in a Büchner ring expansion or cyclopropanation. Mechanistic studies indicated that the spirocyclization occurred through a silver-assisted asynchronous concerted process and not via a silver-carbene intermediate. Analyses based on natural bond orbital population and a distortion/interaction model indicated that the degree of C-Ag mutual interaction is crucial for achieving a high level of enantiocontrol. In addition, an oxidative disconnection of a C(sp3)-C(sp2) bond in the product provided unconventional access to the corresponding chiral spirooxindole.
Collapse
Affiliation(s)
- Jun Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Shingo Harada
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Ayaka Kanda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Hiroki Nakayama
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Tetsuhiro Nemoto
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-8675, Japan.,Molecular Chirality Research Center, Chiba University, 1-33, Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| |
Collapse
|
24
|
Martínez-Castro E, Suárez-Pantiga S, Mendoza A. Scalable Synthesis of Esp and Rhodium(II) Carboxylates from Acetylacetone and RhCl 3· xH 2O. Org Process Res Dev 2020; 24:1207-1212. [PMID: 32587455 PMCID: PMC7309316 DOI: 10.1021/acs.oprd.0c00164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Indexed: 11/28/2022]
Abstract
![]()
Rhodium(II)
carboxylates are privileged catalysts for the most
challenging carbene-, nitrene-, and oxo-transfer reactions. In this
work, we address the strategic challenges of current organic and inorganic
synthesis methods to access these rhodium(II) complexes through an
oxidative rearrangement strategy and a reductive ligation reaction.
These studies illustrate the multiple benefits of oxidative rearrangement
in the process-scale synthesis of congested carboxylates over nitrile
anion alkylation reactions, and the impressive effect of inorganic
additives in the reductive ligation of rhodium(III) salts.
Collapse
Affiliation(s)
- Elisa Martínez-Castro
- Department of Organic Chemistry, Arrhenius Laboratory, Stockholm University, 106 91 Stockholm, Sweden
| | - Samuel Suárez-Pantiga
- Department of Organic Chemistry, Arrhenius Laboratory, Stockholm University, 106 91 Stockholm, Sweden
| | - Abraham Mendoza
- Department of Organic Chemistry, Arrhenius Laboratory, Stockholm University, 106 91 Stockholm, Sweden
| |
Collapse
|
25
|
Hosseini A, Schreiner PR. Synthesis of Exclusively 4-Substituted β-Lactams through the Kinugasa Reaction Utilizing Calcium Carbide. Org Lett 2019; 21:3746-3749. [PMID: 31059273 DOI: 10.1021/acs.orglett.9b01192] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A new Kinugasa reaction protocol has been elaborated for the one-pot synthesis of 4-substituted β-lactams utilizing calcium carbide and nitrone derivatives. Calcium carbide is thereby activated by TBAF·3H2O in the presence of CuCl/NMI. The ease of synthesis and use of inexpensive chemicals provides rapid access of practical quantities of β-lactams exclusively substituted at position 4.
Collapse
Affiliation(s)
- Abolfazl Hosseini
- Institute of Organic Chemistry , Justus Liebig University , Heinrich-Buff-Ring 17 , 35392 Giessen , Germany
| | - Peter R Schreiner
- Institute of Organic Chemistry , Justus Liebig University , Heinrich-Buff-Ring 17 , 35392 Giessen , Germany
| |
Collapse
|
26
|
Pavlinov I, Gerlach EM, Aldrich LN. Next generation diversity-oriented synthesis: a paradigm shift from chemical diversity to biological diversity. Org Biomol Chem 2019; 17:1608-1623. [PMID: 30328455 DOI: 10.1039/c8ob02327a] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Diversity-oriented synthesis adds biological performance as a new diversity element.
Collapse
Affiliation(s)
- Ivan Pavlinov
- University of Illinois at Chicago
- Department of Chemistry
- 845 West Taylor Street
- USA
| | - Erica M. Gerlach
- University of Illinois at Chicago
- Department of Chemistry
- 845 West Taylor Street
- USA
| | - Leslie N. Aldrich
- University of Illinois at Chicago
- Department of Chemistry
- 845 West Taylor Street
- USA
| |
Collapse
|
27
|
Samarium(II) folding cascades involving hydrogen atom transfer for the synthesis of complex polycycles. Nat Commun 2018; 9:4802. [PMID: 30442955 PMCID: PMC6237924 DOI: 10.1038/s41467-018-07194-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 10/16/2018] [Indexed: 01/01/2023] Open
Abstract
The expedient assembly of complex, natural product-like small molecules can deliver new chemical entities with the potential to interact with biological systems and inspire the development of new drugs and probes for biology. Diversity-oriented synthesis is a particularly attractive strategy for the delivery of complex molecules in which the 3-dimensional architecture varies across the collection. Here we describe a folding cascade approach to complex polycyclic systems bearing multiple stereocentres mediated by reductive single electron transfer (SET) from SmI2. Simple, linear substrates undergo three different folding pathways triggered by reductive SET. Two of the radical cascade pathways involve the activation and functionalization of otherwise inert secondary alkyl and benzylic groups by 1,5-hydrogen atom transfer (HAT). Combination of SmI2, a privileged reagent for cascade reactions, and 1,5-HAT can lead to complexity-generating radical sequences that unlock access to diverse structures not readily accessible by other means.
Collapse
|
28
|
|
29
|
Boström J, Brown DG, Young RJ, Keserü GM. Expanding the medicinal chemistry synthetic toolbox. Nat Rev Drug Discov 2018; 17:709-727. [DOI: 10.1038/nrd.2018.116] [Citation(s) in RCA: 267] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
30
|
|
31
|
Davis AM, Plowright AT, Valeur E. Directing evolution: the next revolution in drug discovery? Nat Rev Drug Discov 2017; 16:681-698. [PMID: 28935911 DOI: 10.1038/nrd.2017.146] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The strong biological rationale to pursue challenging drug targets such as protein-protein interactions has stimulated the development of novel screening strategies, such as DNA-encoded libraries, to allow broader areas of chemical space to be searched. There has also been renewed interest in screening natural products, which are the result of evolutionary selection for a function, such as interference with a key signalling pathway of a competing organism. However, recent advances in several areas, such as understanding of the biosynthetic pathways for natural products, synthetic biology and the development of biosensors to detect target molecules, are now providing new opportunities to directly harness evolutionary pressure to identify and optimize compounds with desired bioactivities. Here, we describe innovations in the key components of such strategies and highlight pioneering examples that indicate the potential of the directed-evolution concept. We also discuss the scientific gaps and challenges that remain to be addressed to realize this potential more broadly in drug discovery.
Collapse
Affiliation(s)
- Andrew M Davis
- AstraZeneca R&D Gothenburg, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Alleyn T Plowright
- Integrated Drug Discovery, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Eric Valeur
- AstraZeneca R&D Gothenburg, Pepparedsleden 1, Mölndal, 43150, Sweden
| |
Collapse
|
32
|
Affiliation(s)
- Shiao Y. Chow
- School of Chemistry
and Astbury
Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, U.K
| | - Adam Nelson
- School of Chemistry
and Astbury
Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, U.K
| |
Collapse
|
33
|
Wang B, Huang W, Zhou J, Tang X, Chen Y, Peng C, Han B. Drug design based on pentaerythritol tetranitrate reductase: synthesis and antibacterial activity of Pogostone derivatives. Org Biomol Chem 2017; 15:6548-6556. [DOI: 10.1039/c7ob01429e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We performed molecular docking studies of Pogostone with PETNR and analyzed structure–activity relationships, which guided the structure design and the subsequent facile organocatalytic synthesis of Pogostone derivatives.
Collapse
Affiliation(s)
- Biao Wang
- State Key Laboratory Breeding Base of Systematic Research
- Development and Utilization of Chinese Medicine Resources
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| | - Wei Huang
- Ministry of Education Key Laboratory of Standardization of Chinese Medicine
- School of Pharmacy
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| | - Jin Zhou
- State Key Laboratory Breeding Base of Systematic Research
- Development and Utilization of Chinese Medicine Resources
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| | - Xue Tang
- State Key Laboratory Breeding Base of Systematic Research
- Development and Utilization of Chinese Medicine Resources
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| | - Yang Chen
- Ministry of Education Key Laboratory of Standardization of Chinese Medicine
- School of Pharmacy
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| | - Cheng Peng
- State Key Laboratory Breeding Base of Systematic Research
- Development and Utilization of Chinese Medicine Resources
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| | - Bo Han
- State Key Laboratory Breeding Base of Systematic Research
- Development and Utilization of Chinese Medicine Resources
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| |
Collapse
|
34
|
Chauhan J, Luthra T, Gundla R, Ferraro A, Holzgrabe U, Sen S. A diversity oriented synthesis of natural product inspired molecular libraries. Org Biomol Chem 2017; 15:9108-9120. [DOI: 10.1039/c7ob02230a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Diversity oriented synthesis of natural product inspired compounds from S-tryptophan methyl ester.
Collapse
Affiliation(s)
- Jyoti Chauhan
- Department of Chemistry
- School of Natural Sciences
- Shiv Nadar University
- GautamBudh Nagar
- India
| | - Tania Luthra
- Department of Chemistry
- School of Natural Sciences
- Shiv Nadar University
- GautamBudh Nagar
- India
| | - Rambabu Gundla
- Department of Chemistry
- Gitam Institute of Technology
- GITAM University
- Hyderabad
- India
| | - Antonio Ferraro
- Institute of Pharmacy and Food Chemistry
- University of Würzburg
- Am Hubland
- Germany
| | - Ulrike Holzgrabe
- Institute of Pharmacy and Food Chemistry
- University of Würzburg
- Am Hubland
- Germany
| | - Subhabrata Sen
- Department of Chemistry
- School of Natural Sciences
- Shiv Nadar University
- GautamBudh Nagar
- India
| |
Collapse
|
35
|
Wang ZY, Bi YH, Yang RL, Zhao XJ, Jiang L, Ding CX, Zheng SY. Highly efficient enzymatic synthesis of novel polydatin prodrugs with potential anticancer activity. Process Biochem 2017. [DOI: 10.1016/j.procbio.2016.09.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
36
|
James MJ, O'Brien P, Taylor RJK, Unsworth WP. Selective Synthesis of Six Products from a Single Indolyl α-Diazocarbonyl Precursor. Angew Chem Int Ed Engl 2016; 55:9671-5. [DOI: 10.1002/anie.201605337] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Indexed: 01/19/2023]
Affiliation(s)
| | - Peter O'Brien
- Department of Chemistry; University of York; York YO10 5DD UK
| | | | | |
Collapse
|
37
|
James MJ, O'Brien P, Taylor RJK, Unsworth WP. Selective Synthesis of Six Products from a Single Indolyl α-Diazocarbonyl Precursor. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201605337] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Peter O'Brien
- Department of Chemistry; University of York; York YO10 5DD UK
| | | | | |
Collapse
|
38
|
Bajorath J. Extending accessible chemical space for the identification of novel leads. Expert Opin Drug Discov 2016; 11:825-9. [PMID: 27383145 DOI: 10.1080/17460441.2016.1210126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Jürgen Bajorath
- a Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry , Rheinische Friedrich-Wilhelms-Universität , Bonn , Germany
| |
Collapse
|
39
|
Garcia-Castro M, Zimmermann S, Sankar MG, Kumar K. Gerüstdiversitätsbasierte Synthese und ihre Anwendung bei der Sonden- und Wirkstoffsuche. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201508818] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Miguel Garcia-Castro
- Abteilung Chemische Biologie; Max-Planck-Institut für molekulare Physiologie; Otto-Hahn-Straße 11 44227 Dortmund Deutschland
| | - Stefan Zimmermann
- Abteilung Chemische Biologie; Max-Planck-Institut für molekulare Physiologie; Otto-Hahn-Straße 11 44227 Dortmund Deutschland
| | - Muthukumar G. Sankar
- Abteilung Chemische Biologie; Max-Planck-Institut für molekulare Physiologie; Otto-Hahn-Straße 11 44227 Dortmund Deutschland
| | - Kamal Kumar
- Abteilung Chemische Biologie; Max-Planck-Institut für molekulare Physiologie; Otto-Hahn-Straße 11 44227 Dortmund Deutschland
| |
Collapse
|
40
|
Garcia-Castro M, Zimmermann S, Sankar MG, Kumar K. Scaffold Diversity Synthesis and Its Application in Probe and Drug Discovery. Angew Chem Int Ed Engl 2016; 55:7586-605. [DOI: 10.1002/anie.201508818] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 01/19/2016] [Indexed: 01/19/2023]
Affiliation(s)
- Miguel Garcia-Castro
- Department of Chemical Biology; Max Planck Institute of Molecular Physiology; Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Stefan Zimmermann
- Department of Chemical Biology; Max Planck Institute of Molecular Physiology; Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Muthukumar G. Sankar
- Department of Chemical Biology; Max Planck Institute of Molecular Physiology; Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Kamal Kumar
- Department of Chemical Biology; Max Planck Institute of Molecular Physiology; Otto-Hahn-Strasse 11 44227 Dortmund Germany
| |
Collapse
|
41
|
Rodrigues T, Reker D, Schneider P, Schneider G. Counting on natural products for drug design. Nat Chem 2016; 8:531-41. [PMID: 27219696 DOI: 10.1038/nchem.2479] [Citation(s) in RCA: 775] [Impact Index Per Article: 96.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 02/12/2016] [Indexed: 02/08/2023]
Abstract
Natural products and their molecular frameworks have a long tradition as valuable starting points for medicinal chemistry and drug discovery. Recently, there has been a revitalization of interest in the inclusion of these chemotypes in compound collections for screening and achieving selective target modulation. Here we discuss natural-product-inspired drug discovery with a focus on recent advances in the design of synthetically tractable small molecules that mimic nature's chemistry. We highlight the potential of innovative computational tools in processing structurally complex natural products to predict their macromolecular targets and attempt to forecast the role that natural-product-derived fragments and fragment-like natural products will play in next-generation drug discovery.
Collapse
Affiliation(s)
- Tiago Rodrigues
- Swiss Federal Institute of Technology (ETH), Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| | - Daniel Reker
- Swiss Federal Institute of Technology (ETH), Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| | - Petra Schneider
- Swiss Federal Institute of Technology (ETH), Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland.,inSili.com LLC, Segantinisteig 3, 8049 Zürich, Switzerland
| | - Gisbert Schneider
- Swiss Federal Institute of Technology (ETH), Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| |
Collapse
|
42
|
Karageorgis G, Dow M, Aimon A, Warriner S, Nelson A. Activity-Directed Synthesis with Intermolecular Reactions: Development of a Fragment into a Range of Androgen Receptor Agonists. Angew Chem Int Ed Engl 2015; 54:13538-44. [PMID: 26358926 PMCID: PMC4648041 DOI: 10.1002/anie.201506944] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Indexed: 11/09/2022]
Abstract
Activity-directed synthesis (ADS), a novel discovery approach in which bioactive molecules emerge in parallel with associated syntheses, was exploited to develop a weakly binding fragment into novel androgen receptor agonists. Harnessing promiscuous intermolecular reactions of carbenoid compounds enabled highly efficient exploration of chemical space. Four substrates were prepared, yet exploited in 326 reactions to explore diverse chemical space; guided by bioactivity alone, the products of just nine of the reactions were purified to reveal diverse novel agonists with up to 125-fold improved activity. Remarkably, one agonist stemmed from a novel enantioselective transformation; this is the first time that an asymmetric reaction has been discovered solely on the basis of the biological activity of the product. It was shown that ADS is a significant addition to the lead generation toolkit, enabling the efficient and rapid discovery of novel, yet synthetically accessible, bioactive chemotypes.
Collapse
Affiliation(s)
- George Karageorgis
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of LeedsLeeds, LS2 9JT (UK) E-mail:
| | - Mark Dow
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of LeedsLeeds, LS2 9JT (UK) E-mail:
| | - Anthony Aimon
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of LeedsLeeds, LS2 9JT (UK) E-mail:
| | - Stuart Warriner
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of LeedsLeeds, LS2 9JT (UK) E-mail:
| | - Adam Nelson
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of LeedsLeeds, LS2 9JT (UK) E-mail:
| |
Collapse
|
43
|
Karageorgis G, Dow M, Aimon A, Warriner S, Nelson A. Activity-Directed Synthesis with Intermolecular Reactions: Development of a Fragment into a Range of Androgen Receptor Agonists. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201506944] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
44
|
Zhu C, Xu G, Ding D, Qiu L, Sun J. Copper-Catalyzed Diazo Cross-/Homo-Coupling toward Tetrasubstituted Olefins and Applications on the Synthesis of Maleimide Derivatives. Org Lett 2015; 17:4244-7. [DOI: 10.1021/acs.orglett.5b02037] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Chenghao Zhu
- School of Pharmaceutical Engineering & Life Science, and Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, Changzhou University, Changzhou 213164, P. R. China
| | - Guangyang Xu
- School of Pharmaceutical Engineering & Life Science, and Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, Changzhou University, Changzhou 213164, P. R. China
| | - Dong Ding
- School of Pharmaceutical Engineering & Life Science, and Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, Changzhou University, Changzhou 213164, P. R. China
| | - Lin Qiu
- School of Pharmaceutical Engineering & Life Science, and Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, Changzhou University, Changzhou 213164, P. R. China
| | - Jiangtao Sun
- School of Pharmaceutical Engineering & Life Science, and Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, Changzhou University, Changzhou 213164, P. R. China
| |
Collapse
|
45
|
Regalado EL, Welch CJ. Pushing the speed limit in enantioselective supercritical fluid chromatography. J Sep Sci 2015; 38:2826-32. [DOI: 10.1002/jssc.201500270] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/22/2015] [Accepted: 05/25/2015] [Indexed: 11/05/2022]
Affiliation(s)
- Erik L. Regalado
- Process & Analytical Chemistry; Merck Research Laboratories; Rahway NJ USA
| | | |
Collapse
|
46
|
Kumar S, Kumari R, Pandey R. New insight-guided approaches to detect, cure, prevent and eliminate malaria. PROTOPLASMA 2015; 252:717-753. [PMID: 25323622 DOI: 10.1007/s00709-014-0697-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 09/01/2014] [Indexed: 06/04/2023]
Abstract
New challenges posed by the development of resistance against artemisinin-based combination therapies (ACTs) as well as previous first-line therapies, and the continuing absence of vaccine, have given impetus to research in all areas of malaria control. This review portrays the ongoing progress in several directions of malaria research. The variants of RTS,S and apical membrane antigen 1 (AMA1) are being developed and test adapted as multicomponent and multistage malaria control vaccines, while many other vaccine candidates and methodologies to produce antigens are under experimentation. To track and prevent the spread of artemisinin resistance from Southeast Asia to other parts of the world, rolling circle-enhanced enzyme activity detection (REEAD), a time- and cost-effective malaria diagnosis in field conditions, and a DNA marker associated with artemisinin resistance have become available. Novel mosquito repellents and mosquito trapping and killing techniques much more effective than the prevalent ones are undergoing field testing. Mosquito lines stably infected with their symbiotic wild-type or genetically engineered bacteria that kill sympatric malaria parasites are being constructed and field tested for stopping malaria transmission. A complementary approach being pursued is the addition of ivermectin-like drug molecules to ACTs to cure malaria and kill mosquitoes. Experiments are in progress to eradicate malaria mosquito by making it genetically male sterile. High-throughput screening procedures are being developed and used to discover molecules that possess long in vivo half life and are active against liver and blood stages for the fast cure of malaria symptoms caused by simple or relapsing and drug-sensitive and drug-resistant types of varied malaria parasites, can stop gametocytogenesis and sporogony and could be given in one dose. Target-based antimalarial drug designing has begun. Some of the putative next-generation antimalarials that possess in their scaffold structure several of the desired properties of malaria cure and control are exemplified by OZ439, NITD609, ELQ300 and tafenoquine that are already undergoing clinical trials, and decoquinate, usnic acid, torin-2, ferroquine, WEHI-916, MMV396749 and benzothiophene-type N-myristoyltransferase (NMT) inhibitors, which are candidates for future clinical usage. Among these, NITD609, ELQ300, decoquinate, usnic acid, torin-2 and NMT inhibitors not only cure simple malaria and are prophylactic against simple malaria, but they also cure relapsing malaria.
Collapse
Affiliation(s)
- Sushil Kumar
- SKA Institution for Research, Education and Development (SKAIRED), 4/11 SarvPriya Vihar, New Delhi, 110016, India,
| | | | | |
Collapse
|
47
|
Affiliation(s)
- Derek B Lowe
- Vertex Pharmaceuticals 50 Northern Avenue, Boston, Massachussets 02210, USA
| |
Collapse
|